WO2014001967A1 - Anti-tofacitinib antibodies and uses thereof for drug monitoring - Google Patents

Anti-tofacitinib antibodies and uses thereof for drug monitoring Download PDF

Info

Publication number
WO2014001967A1
WO2014001967A1 PCT/IB2013/055008 IB2013055008W WO2014001967A1 WO 2014001967 A1 WO2014001967 A1 WO 2014001967A1 IB 2013055008 W IB2013055008 W IB 2013055008W WO 2014001967 A1 WO2014001967 A1 WO 2014001967A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
amino acid
tofacitinib
acid sequence
antibody
Prior art date
Application number
PCT/IB2013/055008
Other languages
French (fr)
Inventor
Joseph Paul GARDNER II
Victoria Yatsum WONG
Original Assignee
Pfizer Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to BR112014032916A priority Critical patent/BR112014032916A2/en
Priority to US14/411,356 priority patent/US9408922B2/en
Priority to AU2013282863A priority patent/AU2013282863A1/en
Priority to MX2014015088A priority patent/MX2014015088A/en
Priority to KR1020147036168A priority patent/KR20150014996A/en
Priority to CN201380034143.8A priority patent/CN104411336A/en
Application filed by Pfizer Inc. filed Critical Pfizer Inc.
Priority to RU2014146707A priority patent/RU2014146707A/en
Priority to JP2015519413A priority patent/JP2015527987A/en
Priority to EP13752676.0A priority patent/EP2866840A1/en
Priority to CA2873192A priority patent/CA2873192A1/en
Publication of WO2014001967A1 publication Critical patent/WO2014001967A1/en
Priority to IL236291A priority patent/IL236291A0/en
Priority to HK15108023.7A priority patent/HK1207309A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/646Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the entire peptide or protein drug conjugate elicits an immune response, e.g. conjugate vaccines
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/94Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving narcotics or drugs or pharmaceuticals, neurotransmitters or associated receptors
    • G01N33/9493Immunosupressants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This invention relates to antibodies specific to tofacitinib that selectively bind to tofacitinib over two known metabolites.
  • the selectivity of the anti-tofacitinib antibodies makes them particularly useful for immunoassays.
  • the invention further relates to immunoassays or kits that include antibodies specific for tofacitinib.
  • This invention relates to antibodies specific to tofacitinib, which are useful, e.g., in assay methods and assay kits for monitoring blood levels of the drug.
  • Tofacitinib is a potent immunosuppressant in development for the treatment of rheumatoid arthritis (RA), psoriasis, inflammatory bowel disease, and other immunological diseases, as well as for the prevention of organ transplant rejection.
  • Tofacitinib specifically inhibits Janus activated kinase 3 (JAK3), which has a pivotal role in cytokine signal transduction governing lymphocyte survival, proliferation, differentiation and apoptosis.
  • JNK3 Janus activated kinase 3
  • organs such as kidney, heart, lung, bone marrow, and liver are transplanted in humans
  • the body will sometimes reject the transplanted tissue.
  • One treatment of organ transplant rejection comprises suppression of the immune system in a controlled manner with immunosuppressant drugs such as tofacitinib.
  • Immunosuppressant drugs are carefully administered to transplant recipients to prevent rejection of the foreign (i.e. non-self) tissue.
  • Successful treatment with immunosuppressant drugs requires the measurement of drug concentrations, followed by subsequent dosage adjustments to maximize efficacy while minimizing toxicity. Monitoring blood levels of tofacitinib in patients treated with tofacitinib is thus very desirable in order to regulate the dosage.
  • TDM Therapeutic Drug Monitoring
  • LC-MS/MS liquid chromatography-mass spectrometry
  • anti-tofacitinib antibodies that bind tofacitinib but do not substantially bind at least one metabolite of tofacitinib. Such antibodies would be useful, among other things, to detect clinically relevant concentrations of tofacitinib in a sample from a patient undergoing therapy wherein tofacitinib is administered to the patient.
  • the present invention is directed to novel immunogenic tofacitinib conjugates, as well as labeled tofacitinib competitors.
  • the present invention is also directed to polyclonal and monoclonal antibodies generated using the immunogenic tofacitinib conjugates. These antibodies, conjugates, and competitors are useful in, among other things, immunoassays for the detection of tofacitinib in a sample.
  • the present invention includes an immunogenic tofacitinib conjugate comprising tofacitinib coupled to an immunogenic carrier, wherein the immunogenic carrier is a protein selected from the group consisting of keyhole limpet hemocyanin and bovine serum albumin, and further wherein
  • BSA bovine serum albumin
  • KLH is keyhole limpet hemocyanin.
  • the invention includes a method for assessing the concentration of tofacitinib in a sample.
  • the method comprises:
  • the invention includes an isolated antibody having greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
  • the isolated antibody has a binding affinity for tofacitinib that is 25, 30, 40, or 50 times the binding affinity for metabolite 1 or metabolite 2.
  • the antibody is obtained using an immunogenic tofacitinib conjugate compound comprising tofacitinib coupled to an immunogenic carrier.
  • the invention includes a method of determining the amount of tofacitinib in a sample.
  • the method comprises:
  • the selective anti-tofacitinib antibody has a greater binding affinity for t bolite 1 of Formula II
  • the antibody has greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
  • the invention includes a kit for determining the amount of tofacitinib in a sample.
  • the kit comprises:
  • a labeled competitor comprising tofacitinib coupled to a detectable label; and at least one selective anti-tofacitinib antibody; wherein the labeled competitor competes with the tofacitinib in a sample for binding to the anti-tofacitinib antibody.
  • the selective anti-tofacitinib antibody has a greater binding affinity for tofacitinib than for at least one tofacitinib metabolite selected from the group consisting of tofacitinib metabolite 1 of Formula II
  • the antibody has greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
  • the kit can be used to detect tofacitinib in a sample at a concentration ranging from about 5 ng/ml to about 1215 ng/ml and comprises
  • an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24; and/or
  • an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
  • the invention includes a competitive immunoassay kit for determining the concentration of tofacitinib in a sample.
  • the competitive immunoassay comprises: at least one selective anti-tofacitinib antibody; a tofacitinib compound conjugated to a detectable label; wherein the conjugated tofacitinib compound competes with the tofacitinib in the sample to bind with the antibody; and wherein the label provides a signal indicative of the concentration of tofacitinib in the sample when the tofacitinib in the sample is present in a therapeutic drug monitoring concentration.
  • the antibody has greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
  • the invention includes an isolated antibody that binds to tofacitinib but does not substantially bind to at least one tofacitinib metabolite selected from the group consisting of a tofacitinib metabolite 1 of Formula II and a tofacitinib metabolite 2 of Formula III
  • the isolated antibody does not substantially bind a tofacitinib metabolite 1 of Formula II and a tofacitinib metabolite 2 of Formula I II
  • the antibody can detect tofacitinib in a sample but not substantially detect a tofacitinib metabolite in the sample, and wherein the amount of tofacitinib ranges from about 5 ng/mL to 1215 ng/mL, from about 5 ng/mL to 405 ng/mL, or about 15 ng/mL to 1215 ng/mL.
  • the invention includes an isolated antibody having greater binding affinity for tofacitinib than for at least one tofacitinib metabolite selected from the group consisting of a tofacitinib metabolite 1 of Formula II and a tofacitinib metabolite 2 of Formula III.
  • the antibody comprises a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain comprises three complementarity determining regions (HCDR1 , HCDR2, and HCDR3) selected from the group consisting of:
  • a HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:13, SEQ ID NO:19, SEQ ID NO:30, and SEQ ID NO:37;
  • a HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:14, SEQ ID NO:20, SEQ ID NO:25, SEQ ID NO:31 , and SEQ ID NO:38;
  • a HCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:15, SEQ ID NO:21 , SEQ ID NO:26, SEQ ID NO:32, and SEQ ID NO:39; and wherein the light chain variable domain comprises three CDRs (LCDR1 , LCDR2, and LCDR3) selected from the group consisting of:
  • a LCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:16, SEQ ID NO:22, SEQ ID NO:27, SEQ ID NO:33, and SEQ ID NO:34;
  • a LCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:17, SEQ ID NO:23, SEQ ID NO:28, and SEQ ID NO:35;
  • a LCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:18, SEQ ID NO:24, SEQ ID NO:29, and SEQ ID NO:34.
  • the antibody comprises
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:13, a HCDR2 comprising the amino acid sequence of SEQ ID NO:14, a HCDR3 comprising the amino acid sequence of SEQ ID NO:15, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:16, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:25, a HCDR3 comprising the amino acid sequence of SEQ ID NO:26, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:27, a LCDR2 comprising the amino acid sequence of SEQ ID NO:28, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:29;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:33, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:34, a LCDR2 comprising the amino acid sequence of SEQ ID NO:35, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:36;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
  • a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:10 and SEQ ID NO:12, and further comprising a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:9, and SEQ ID NO:1 1 ;
  • a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID N03, SEQ ID NO:7 and SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
  • a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:8, SEQ ID NO:10, and SEQ ID NO:12 and a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:4, SEQ ID NO:9 and SEQ ID NO:1 1.
  • the antibody is selected from the group consisting of:
  • an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24:
  • an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
  • an antibody comprising a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:7 and SEQ ID NO:12 and further comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
  • an antibody comprising a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and further comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
  • an antibody comprising a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:12 and further comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4.
  • the antibody is capable of detecting tofacitinib in a sample at a concentration ranging from about 15 ng/ml to 1215 ng/ml.
  • the antibody is capable of detecting tofacitinib in a sample at a concentration ranging from about 5 ng/ml to 405 ng/ml.
  • the invention includes a nucleic acid encoding an antibody comprising
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:13, a HCDR2 comprising the amino acid sequence of SEQ ID NO:14, a HCDR3 comprising the amino acid sequence of SEQ ID NO: 15, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO: 16, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:25, a HCDR3 comprising the amino acid sequence of SEQ ID NO:26, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:27, a LCDR2 comprising the amino acid sequence of SEQ ID NO:28, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:29;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:33, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18; (f) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:34, a LCDR2 comprising the amino acid sequence of SEQ ID NO:35, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:36;
  • a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
  • a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:10 and SEQ ID NO:12, and further comprising a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:9, and SEQ ID NO:1 1 ;
  • a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:10 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:1 1 ;
  • a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID N03, SEQ ID NO:7 and SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
  • a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:8, SEQ ID NO:10, and SEQ ID NO:12 and a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:4, SEQ ID NO:9 and SEQ ID NO:1 1.
  • the invention includes a host cell comprising the nucleic acid.
  • the invention includes a method of producing the antibody.
  • the method comprises culturing the host cell under conditions wherein the antibody is produced.
  • the method comprises isolating the antibody.
  • Figure 1 is a graph depicting binding of serum antibodies to the tofacitinib-BSA conjugate of example 2 as determined by solid phase ELISA.
  • Figure 2 is a graph depicting binding of selected anti-tofacitinib antibodies to biotinylated tofacitinib as determined by ELISA.
  • Figure 3 is a graph depicting cross reactivity of selected anti-tofacitinib antibodies to metabolite 1 as determined by ELISA.
  • Figure 4 is a graph depicting optimal dilution ranges of select monoclonal antibodies of the invention as determined by ELISA.
  • Figure 5 is a graph depicting optimal dilution ranges of select monoclonal antibodies of the invention as determined by ELISA.
  • the present invention provides novel immunogenic tofacitinib conjugates comprising tofacitinib coupled to an immunogenic carrier, as well as labeled tofacitinib competitors.
  • the present invention is also directed to polyclonal and monoclonal antibodies generated using the immunogenic tofacitinib conjugates.
  • the present invention also provides polyclonal and monoclonal antibodies which are specific for tofacitinib.
  • the antibodies of the invention are produced in response to inoculation with a novel immunogenic tofacitinib conjugate comprising tofacitinib linked to an immunogenic carrier.
  • These antibodies, conjugates, and competitors are useful in immunoassays for the detection of tofacitinib in biological fluids.
  • Assay kits comprising these antibodies, conjugates and competitors are well suited for use in a clinical setting and provide far more accurate and reproducible results than was previously possible.
  • the antibodies are also useful in the purification and isolation of tofacitinib.
  • antibody as referred to herein includes whole antibodies and any antigen binding fragment (i.e., "antigen-binding portion") or single chains thereof.
  • An “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof.
  • An “antibody” also refers to an IgA, IgD, IgE, IgG, IgM antibody subtype.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as V H ) and a heavy chain constant region (C H ).
  • the heavy chain constant region is typically comprised of three domains, Cm, C H 2 and C H 3-
  • Each light chain is comprised of a light chain variable region (abbreviated herein as V L ) and a light chain constant region.
  • the light chain constant region is comprised of one domain, C L .
  • the V H and V L regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each V H and V L is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin
  • a "CDR" of a variable domain are amino acid residues within the variable region that are identified in accordance with the definitions of the Kabat, Chothia, the accumulation of both Kabat and Chothia, AbM, contact, and/or conformational definitions or any method of CDR determination well known in the art.
  • Antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. See, e.g., Kabat et al., 1992,
  • CDRs Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, NIH, Washington D.C.
  • the positions of the CDRs may also be identified as the structural loop structures originally described by Chothia and others. See, e.g., Chothia et al., 1989, Nature 342:877-883.
  • Other approaches to CDR identification include the "IMGT definition" (Lefranc, M.-P. et al., 1999, Nucleic Acids Res.
  • CDR boundary definitions may not strictly follow one of the above approaches, but will nonetheless overlap with at least a portion of the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding.
  • a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches. The methods used herein may utilize CDRs defined according to any of these approaches. For any given embodiment containing more than one CDR, the CDRs may be defined in accordance with any of Kabat, Chothia, extended, AbM, contact, and/or conformational definitions.
  • antigen-binding portion or "antigen-binding fragment” of an antibody (or simply “antibody portion”), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., tofacitinib). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full- length antibody.
  • binding fragments encompassed within the term "antigen- binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the V
  • a Fab fragment a monovalent fragment consisting of the V
  • a F(ab')2 fragment a bivalent fragment comprising two Fab fragment
  • the two domains of the Fv fragment, V L and V H are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the V L and V H regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al., 1988, Science 242:423-426; and Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen- binding portion" of an antibody.
  • an "isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds tofacitinib is substantially free of antibodies that specifically bind antigens other than tofacitinib). Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.
  • monoclonal antibody or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition.
  • a monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
  • recombinant antibody includes all antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from a host cell transformed to express the antibody, (b) antibodies isolated from a recombinant, combinatorial antibody library, and (c) antibodies prepared, expressed, created or isolated by any other means that involve splicing of immunoglobulin gene sequences to other DNA sequences.
  • recombinant antibodies comprise variable regions in which the framework and CDR regions are derived from mouse germline immunoglobulin sequences.
  • such recombinant mouse antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the V H and V L regions of the recombinant antibodies are sequences that, while derived from and related to mouse germline V H and V L sequences, may not naturally exist within the mouse antibody germline repertoire in vivo.
  • isotype refers to the antibody class (e.g., IgM or IgG) that is encoded by the heavy chain constant region genes.
  • an antibody recognizing an antigen and "an antibody specific for an antigen” are used interchangeably herein with the term “an antibody which binds specifically to an antigen.”
  • antibody derivatives refers to any modified form of the antibody, e.g., a conjugate of the antibody and another agent or antibody.
  • humanized antibody is intended to refer to antibodies in which CDR sequences derived from the germline of a non-human species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications may be made within the human framework sequences.
  • chimeric antibody is intended to refer to antibodies in which the variable region sequences are derived from one species and the constant region sequences are derived from another species, such as an antibody in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody. Chimeric antibody can also include an antibody where the V domain and C domain are each derived two different sources even if both are from the same species.
  • an antibody that "specifically binds to tofacitinib" or "a selective tofacitinib antibody” refers to an antibody that binds to tofacitinib but which does not substantially bind a metabolite of tofacitinib.
  • An antibody specifically binds tofacitinib where it does not detectibly binds to a tofacitinib metabolite 1 of Formula II and/or to a tofacitinib metabolite 2 of Formula III or binds to such metabolite to a much lesser extent.
  • the metabolite does not substantially compete with tofacitinib for binding to the antibody as measured by, for example, a competitive binding assay.
  • a selective tofacitinib antibody binds to tofacitinib with an affinity that is 5, 10, 15, 25, 30, 40, or 50 times greater than its affinity to metabolite 1 of Formula II or metabolite 2 of Formula III.
  • the term “subject” includes any human or nonhuman animal.
  • nonhuman animal includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows chickens, amphibians, reptiles, etc.
  • variable region of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination.
  • variable regions of the heavy and light chain each consist of four framework regions (FRs) connected by three complementarity determining regions (CDRs) also known as hypervariable regions, contribute to the formation of the antigen binding site of antibodies.
  • FRs framework regions
  • CDRs complementarity determining regions
  • variants of a subject variable region are desired, particularly with substitution in amino acid residues outside of a CDR region (i.e., in the framework region)
  • appropriate amino acid substitution preferably, conservative amino acid substitution
  • FR to flank subject CDRs e.g., when humanizing or optimizing an antibody, FRs from antibodies which contain CDR1 and CDR2 sequences in the same canonical class are preferred.
  • a "CDR" of a variable domain are amino acid residues within the variable region that are identified in accordance with the definitions of the Kabat, Chothia, the accumulation of both Kabat and Chothia, AbM, contact, and/or conformational definitions or any method of CDR determination well known in the art.
  • Antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. See, e.g., Kabat et al., 1992, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, NIH, Washington D.C. The positions of the CDRs may also be identified as the structural loop structures originally described by Chothia and others.
  • CDR identification See, e.g., Chothia et al., 1989, Nature 342:877-883.
  • Other approaches to CDR identification include the "IMGT definition” (Lefranc, M.-P. et al., Nucleic Acids Res., 27, 209-212 (1999)) and the “AbM definition,” which is a compromise between Kabat and Chothia and is derived using Oxford Molecular's AbM antibody modeling software (now Accelrys®), or the "contact definition" of CDRs based on observed antigen contacts, set forth in MacCallum et al., 1996, J. Mol. Biol., 262:732-745.
  • CDRs In another approach, referred to herein as the "conformational definition" of CDRs, the positions of the CDRs may be identified as the residues that make enthalpic contributions to antigen binding. See, e.g., Makabe et al., 2008, Journal of Biological Chemistry, 283:1 156- 1 166. Still other CDR boundary definitions may not strictly follow one of the above approaches, but will nonetheless overlap with at least a portion of the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. As used herein, a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches.
  • CDRs defined according to any of these approaches.
  • the CDRs may be defined in accordance with any of Kabat, Chothia, extended, AbM, contact, and/or conformational definitions.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single copy or clone, including e.g., any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
  • a monoclonal antibody of the invention exists in a homogeneous or substantially homogeneous population.
  • Antibodies of the invention can be produced using techniques well known in the art, e.g., recombinant technologies, phage display technologies, synthetic technologies or combinations of such technologies or other technologies readily known in the art (see, for example, Jayasena, S.D., 1999, Clin. Chem. 45:1628-1650 and Fellouse et al., 2007, J. Mol. Biol., 373(4):924-940).
  • binding refers to the ability to bind to a target compound or epitope with greater affinity than to a non-target compound.
  • an antibody that specifically binds tofacitinib has a greater affinity for tofacitinib than for metabolites of tofacitinib, e.g., metabolite 1 of Formula II and metabolite 2 of Formula III.
  • “specifically binds” or “selectively binds” refers to binding to a target compound with an affinity that is at least 5, 10, 15, 20, 25, 30, 50, 100, 500, 1000 or more times greater than the affinity for a non-target.
  • epitope refers to that portion of a molecule capable of being recognized by and bound by an antibody at one or more of the antibody's antigen-binding regions. Epitopes often consist of a chemically active surface grouping of molecules such as amino acids or sugar side chains and have specific three-dimensional structural characteristics as well as specific charge characteristics.
  • antigenic epitope as used herein, is defined as a portion of a polypeptide to which an antibody can specifically bind as determined by any method well known in the art, for example, by conventional immunoassays.
  • a “nonlinear epitope” or “conformational epitope” comprises noncontiguous polypeptides (or amino acids) within the antigenic protein to which an antibody specific to the epitope binds.
  • an immunogen can also be antigen.
  • an immunogen has a fairly high molecular weight (e.g., greater than 10,000), thus, a variety of macromolecules such as proteins, lipoproteins, polysaccharides, some nucleic acids, and certain of the teichoic acids, can be an immunogen.
  • Haptens are partial or incomplete antigens. They are usually protein-free substances, mostly of low molecular weight, which are not generally capable of stimulating antibody formation, but which do react with antibodies. Antibodies to a hapten may be generated by coupling a hapten to a high molecular weight antigenic carrier (e.g., an immunogen) and then injecting this coupled product, i.e., an immunogenic conjugate, into a human or animal subject.
  • a high molecular weight antigenic carrier e.g., an immunogen
  • tofacitinib is a hapten.
  • a “carrier” or “immunogenic carrier,” as the terms are used herein, is an immunogenic substance, commonly a protein, that can join with a hapten, thereby enabling the hapten to induce an immune response and elicit the production of antibodies that can bind specifically with the antigen (hapten).
  • Carrier substances include proteins, glycoproteins, complex polysaccharides, particles, and nucleic acid that are recognized as foreign and thereby elicit an immunologic response from the host.
  • proteins may be employed as a polypeptide immunogenic carrier. These proteins include albumins and serum proteins, e.g., globulins, ocular lens proteins, lipoproteins, etc.
  • Illustrative proteins include bovine serum albumin (BSA), keyhole limpet hemocyanin (KLH), egg ovalbumin, bovine gamma-globulin (BGG), etc.
  • BSA bovine serum albumin
  • KLH keyhole limpet hemocyanin
  • BGG bovine gamma-globulin
  • synthetic polypeptides may be used.
  • the immunogenic carrier can also be a polysaccharide such as, e.g., starches, glycogen, cellulose, carbohydrate gums such as gum arabic, agar, and so forth.
  • the polysaccharide can also contain polypeptide residues and/or lipid residues.
  • the immunogenic carrier can also be a polynucleotide either alone or conjugated to one of the polypeptides or polysaccharides mentioned above.
  • the term "immunogenicity” is meant to refer to the ability of a molecule to induce an immune response, which may be determined both by the intrinsic chemical structure of the injected molecule and by whether or not the host animal's immune system recognizes the compound.
  • Small changes in the structure of an antigen can greatly alter the immunogenicity of a compound, and have been used extensively as a general procedure to increase the chances of raising an antibody, particularly against well-conserved antigens. For example, these modification techniques either alter regions of the immunogen to provide better sites for T-cell binding or expose new epitopes for B-cell binding.
  • label refers to any molecule that produces, or can be induced to produce, a detectable signal.
  • the label can be conjugated to an analyte, an immunogen, and/or an antibody.
  • Non-limiting examples of labels include radioactive isotopes, enzymes, enzyme fragments, enzyme substrates, enzyme inhibitors, coenzymes, catalysts, fluorophores, dyes, chemiluminescers, luminescers, sensitizers, non-magnetic or magnetic particles, solid supports, liposomes, ligands, receptors, and hapten radioactive isotopes.
  • labeled with regard to a specific antibody or a labeled competitor, includes direct labeling by coupling (i.e., physically linking) a detectable substance to the antibody or labeled competitor, as well as indirect labeling of the antibody or labeled competitor by coupling it with another reagent that is, in turn, directly labeled.
  • indirect labeling includes detection of a primary antibody using a fluorescent-labeled secondary antibody.
  • In vitro techniques for detection of an antigen of the invention include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitation, and immunofluorescence.
  • the antibodies, labeled competitors, and potential therapeutic compounds described herein are also suitable for use with any of a number of other homogeneous and heterogeneous immunoassays with a range of detection systems.
  • the term "antigenic compound” refers to a compound used to produce an immune response.
  • the antigenic compound is a hapten, for example tofacitinib, linked to an immunogenic carrier.
  • the antigenic compound is used to generate the desired antibodies.
  • labeled competitor is a molecule capable of specific binding to antibodies having specificity for tofacitinib, wherein the molecule is linked to a detectable label or tracer.
  • the molecule is tofacitinib or a derivative or analyte thereof.
  • biological sample includes, but is not limited to, any quantity of a substance isolated or derived from a living subject or formerly living subject.
  • the term is intended to include tissues, cells, and biological fluids isolated from a subject, as well as tissues, cells, and fluids present within a subject.
  • Subjects include, but are not limited to, chicken, humans, mice, monkeys, rats, rabbits, horses, camelids, and other animals.
  • Such substance include, but are not limited to, blood, plasma, serum, semen, urine, tears, saliva, cells, organs, tissues, bone, bone marrow, lymph, lymph nodes, synovial tissue or fluid, chondrocytes, synovial macrophages, endothelial cells, and skin.
  • this disclosure provides derivatives of tofacitinib which are useful as immunogenic molecules for the generation of antibodies specific for tofacitinib for measuring levels of tofacitinib.
  • Tofacitinib (Formula I) is an orally available, potent selective inhibitor of JAK3 that is in development for the treatment of rheumatoid arthritis (RA) and other autoimmune disorders, such as inflammatory bowel disease, ankylosing spondylitis, psoriasis, psoriatic arthritis, and the prevention of transplant rejection.
  • RA rheumatoid arthritis
  • JAK3 rheumatoid arthritis
  • other autoimmune disorders such as inflammatory bowel disease, ankylosing spondylitis, psoriasis, psoriatic arthritis, and the prevention of transplant rejection.
  • an immunoassay for the detection of a small molecule such as tofacitinib
  • small molecules lack antigenicity, making it difficult to generate antibodies against them. This challenge is compounded for tofacitinib because it has such potent immunosuppressive properties.
  • larger immunogenic compounds including but not limited to bovine serum albumin, ovalbumin, keyhole limpet hemocyanin, and the like, can be coupled to the drug. Detection of the drug in a sample generally requires the use of a detectable label conjugated to an antibody, tofacitinib, or tofacitinib analog.
  • this disclosure provides an immunogenic tofacitinib conjugate comprising tofacitinib coupled to an immunogenic carrier, wherein the immunogenic carrier is a protein, glycoprotein, complex polysaccharide, or nucleic acid that is recognized by the immunized animal as foreign, resulting in an immunologic response.
  • the immunogenic carrier is a protein selected from the group consisting of keyhole limpet hemocyanin (KLH) and bovine serum albumin (BSA).
  • this disclosure provides an immunogenic tofacitinib conjugate of formula V, having the structure
  • BSA bovine serum albumin
  • this disclosure provides an immunogenic tofacitinib conjugate of formula VI, having the structure
  • KLH Keyhole Limpet Hemocyanin
  • the immunogenic tofacitinib conjugates of the present invention can be used to elicit antibodies using known antibody producing and screening procedures.
  • the immunogenic tofacitinib conjugates can be injected into appropriate animal hosts to stimulate the production of antibodies.
  • the antibodies so produced can be harvested for direct use in immunoassays configured to detect tofacitinib.
  • the antibody has a binding affinity for tofacitinib that is about 25, 30, 40, or 50 times the binding affinity for metabolite 1 or metabolite 2.
  • the antibody binds to metabolite 1 or metabolite 2 with an affinity that is less than about 5%, 4%, 3%, 2%, 1 %, or 0.5% of the binding affinity to tofacitinib.
  • the antibodies of the present invention are characterized by the fact that they bind specifically to tofacitinib (Formula I), but which do not substantially bind to one or both known tofacitinib metabolites, "metabolite 1 of Formula II” and “metabolite 2 of Formula III,” and methods of making and using these anti-tofacitinib antibodies.
  • a selective tofacitinib antibody binds to tofacitinib with an affinity that is 5, 10, 15, 25, 30, 40, or 50 times greater than its affinity to metabolite 1 of Formula II or metabolite 2 of Formula III.
  • Standard assays to evaluate the binding ability of the antibodies toward tofacitinib and its metabolites are known in the art, including for example, ELISAs, Western blots, radioimmunoassays, and flow cytometry analysis. Suitable assays are described in detail in the Examples.
  • the binding kinetics (e.g., binding affinity) of the antibodies also can be assessed by standard assays known in the art, such as by Biacore SPR analysis and Octet analysis.
  • the antibodies of the invention include mouse monoclonal antibodies 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6.
  • the V H amino acid sequences of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in Table 2, and are set forth in SEQ ID NOs: 1 , 3, 5, 7, 8, 10 and 12, respectively.
  • V L amino acid sequences of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in Table 2 and are set forth in SEQ ID NOs: 2, 4, 6, 4, 9, 1 1 and 4, respectively.
  • V H and V L sequences may be "mixed and matched" to create other selective tofacitinib binding molecules of the invention. Tofacitinib binding of such mixed and matched antibodies, as well as the binding of the antibody to one or both metabolites 1 and 2, can be tested using the binding assays described above and in the Examples.
  • V H and V L chains are mixed and matched, a V H sequence from a particular V H A L pairing is replaced with a structurally similar V H sequence.
  • a V L sequence from a particular V H A L pairing is replaced with a structurally similar V L sequence.
  • the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising:
  • a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1 , SEQ I D NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:10 and SEQ ID NO:12, and further comprising a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2, SEQ I D NO:4, SEQ ID NO:6, SEQ ID NO:9, and SEQ ID NO:1 1 ; (b) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:1 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:2;
  • a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID N03, SEQ ID NO:7 and SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
  • a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ I D NO:8, SEQ ID NO:10, and SEQ ID NO:12 and a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:4, SEQ ID NO:9 and SEQ ID NO:1 1 .
  • the invention provides antibodies that comprise the heavy chain and/or light chain CDR1 s, CDR2s and CDR3s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6, or combinations thereof.
  • the amino acid sequences of the V H CDR1 S of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs: 13, 19, 19, 30, 19, 19, and 37, respectively.
  • the amino acid sequences of the V H CDR2s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs: 14, 20, 25, 31 , 20, 20, and 38, respectively.
  • the amino acid sequences of the V H CDR3s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs:15, 21 , 26, 32, 21 , 21 , and 39, respectively.
  • the amino acid sequences of the V L CDR1 s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs:16, 22, 27, 22, 33, 34, and 22, respectively.
  • the amino acid sequences of the V L CDR2s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs:17, 23, 28, 23, 17, 35 and 23, respectively.
  • the amino acid sequences of the V L CDR3s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs:18, 24, 29, 24, 18, 36, and 24, respectively.
  • the CDR regions are delineated using the Kabat system (Kabat, E. A., et al. Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242 (1991 )).
  • the heavy and light chain CDRs for the antibody 5A3.E5 comprise SEQ ID NOs: 13, 14, 15, 16, 17, 18.
  • the heavy and light chain CDRs for the antibody 10A6.C5 comprise SEQ ID NOs: 19, 20, 21 , 22, 23, and 24.
  • the heavy and light chain CDRs for the antibody 10F10.H5 comprise SEQ ID NOs:19, 25, 26, 27, 28, and 29.
  • the heavy and light chain CDRs for the antibody 6D9.A5 comprise SEQ ID NOs:30, 31 , 32, 22, 23, and 24.
  • the heavy and light chain CDRs for the antibody 12H4.G2 comprise SEQ ID NOs:19, 20, 21 , 33, 17, and 18.
  • the heavy and light chain CDRs for the antibody 16F10.E6 comprise SEQ ID NOs:19, 20, 21 , 34, 35, and 36.
  • the heavy and light chain CDRs for the antibody 12D4.G6 comprise SEQ ID NOs:37, 38, 39, 22, 23, and 24.
  • an antibody of the invention comprises heavy and light chain variable regions and/or heavy chain and light chain CDR1 s, CDR2s and CDR3s comprising amino acid sequences that are homologous to the amino acid sequences of the antibodies previously described herein, and wherein the antibodies retain the desired functional properties of the selective anti-tofacitinib antibodies of the invention.
  • the antibody can be, for example, a mouse antibody, a humanized antibody or a chimeric antibody derived from a mouse anti-tofacitinib antibody.
  • the V H and/or V L amino acid sequences may be 85%, 90%, 95%, 96%, 97%, 98% or 99% homologous to the sequences set forth above.
  • the percent homology between two amino acid sequences is equivalent to the percent identity between the two sequences.
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • an antibody of the invention comprises heavy and light chain variable regions and/or heavy chain and light chain CDR1 s, CDR2s and CDR3s, wherein one or more of these sequences comprise specified amino acid sequences based on the antibodies described herein, or conservative modifications thereof, and wherein the antibodies retain the desired functional properties of the anti-tofacitinib antibodies of the invention.
  • the antibody can be, for example, mouse antibodies, humanized antibodies or chimeric antibodies.
  • conservative sequence modifications is intended to refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody of the invention by standard techniques known in the art, such as site- directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • the invention provides an antibody that binds to the same epitope on tofacitinib as the anti-tofacitinib antibodies of the invention (i.e., an antibody that has the ability to compete for binding to tofacitinib with an antibody of the invention).
  • the reference antibody for competition studies can be the monoclonal antibody 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6.
  • Such competing antibodies can be identified based on their ability to compete with 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, or 12D4.G6 for binding of tofacitinib in standard binding assays.
  • Biacore analysis, Octect analysis, ELISA assays or flow cytometry may be used to demonstrate cross-competition with the antibodies of the current invention.
  • test antibody to inhibit the binding of, for example, 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, or 12D4.G6, to tofacitinib, wherein the test antibody does not substantially bind metabolite 1 and/or metabolite 2, demonstrates that the test antibody can compete with 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 for binding to tofacitinib and thus may bind the same epitope on tofacitinib as 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6.
  • the antibody that binds to the same epitope on tofacitinib as 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 is a mouse monoclonal antibody.
  • Such monoclonal antibodies can be prepared and isolated as described in the Examples or by a wide variety of methods well-known in the art.
  • the antibody that competes with the antibody is a human, humanized or mouse antibody.
  • An antibody of the invention can be prepared using an antibody comprising at least one of the V H and/or V L sequence disclosed herein as starting material to engineer a modified antibody, which may have properties that differ from the starting antibody but which may bind the same, or substantially the same, epitope as the starting material antibody.
  • An antibody can be engineered by modifying one or more residues within one or both variable regions (i.e., V H and/or V L ), for example within one or more CDR regions and/or within one or more framework regions. Additionally or alternatively, an antibody can be engineered by modifying residues within the constant region(s), for example to alter the effector function(s) of the antibody.
  • the present invention provides nucleic acids encoding the tofacitinib specific antibody of the invention.
  • Nucleic acids encoding the antibodies of the invention can be generated by methods known in the art. Also, as would be understood by one skilled in the art, due to the degeneracy of the nucleic acid code, a wide variety of nucleic acid sequences can encode the amino acid sequence of the antibody of the invention.
  • CDR grafting One type of variable region engineering that can be performed is CDR grafting. Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs.
  • CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of specific naturally occurring antibodies by constructing expression vectors that include CDR sequences from the specific naturally occurring antibody grafted onto framework sequences from a different antibody with different properties (see, e.g., Riechmann et al., 1998, Nature 332:323-327; Jones et al., 1986, Nature 321 :522-525; Queen et al., 1989, Proc. Natl. Acad. See. U.S.A. 86:10029-10033; U.S. Patent Nos. 5,225,539; 5,530,101 ; 5,585,089; 5,693,762 and 6,180,370).
  • another embodiment of the invention pertains to an isolated monoclonal antibody, or antigen binding portion thereof, comprising a heavy chain variable region comprising CDR1 , CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 13, 19, 30, and 37; SEQ ID NOs:14, 20, 25, 31 , and 38; and SEQ ID NOs:15, 21 , 26, 32, and 39, respectively, and a light chain variable region comprising CDR1 , CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:16, 22, 27, 33, and 34; SEQ ID NOs: 17, 23, 28, and 35; and SEQ ID NOs:18, 24, 29, and 36, respectively.
  • Such antibodies contain the V H and V L CDR sequences of monoclonal antibodies 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6, which may contain a different framework sequence from the antibodies.
  • Such framework sequences can be obtained from public DNA databases or published references that include germline antibody gene sequences.
  • variable region modification is to mutate amino acid residues within the V H and/or V L CDR1 , CDR2 and/or CDR3 regions to thereby improve one or more binding properties (e.g., affinity) of the antibody of interest.
  • Site-directed mutagenesis or PCR- mediated mutagenesis can be performed to introduce the mutation(s) and the effect on antibody binding, or other functional property of interest, can be evaluated in in vitro or in vivo assays as described herein and provided in the Examples.
  • conservative modifications are introduced.
  • the mutations may be amino acid substitutions, additions or deletions.
  • typically no more than one, two, three, four or five residues within a CDR region are altered.
  • the invention provides isolated anti-phospho-tau monoclonal antibodies, or antigen binding portions thereof, comprising a heavy chain variable region comprising:
  • V H CDR1 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:13, 19, 30, and 37, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 13, 19, 30, and 37
  • V H CDR2 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 14, 20, 25, 31 , and 38, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 14, 20, 25, 31 , and 38
  • V H CDR3 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 15, 21 , 26, 32, and 39, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 15, 21 , 26, 32, and 39
  • V L CDR3 region comprising an amino acid sequence selected from the group consisting of S
  • Engineered antibodies of the invention include those in which modifications have been made to framework residues within V H and/or V L , e.g., to improve the properties of the antibody. Typically such framework modifications are made to decrease the immunogenicity of the antibody. For example, one approach is to "backmutate" one or more framework residues to the corresponding germline sequence (also referred to as "germlining"). More specifically, an antibody that has undergone somatic mutation may contain framework residues that differ from the germline sequence from which the antibody is derived. Such residues can be identified by comparing the antibody framework sequences to the germline sequences from which the antibody is derived.
  • Another type of framework modification involves mutating one or more residues within the framework region, or even within one or more CDR regions, to remove T cell epitopes to thereby reduce the potential immunogenicity of the antibody. This approach is also referred to as “deimmunization” and is described in further detail in U.S. Patent
  • antibodies of the invention may be engineered to include modifications within the Fc region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen- dependent cellular cytotoxicity.
  • modifications within the Fc region typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen- dependent cellular cytotoxicity.
  • an antibody of the invention may be chemically modified (e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation.
  • An antibody can be pegylated to, for example, increase the biological (e.g., serum) half life of the antibody.
  • the antibody, or fragment thereof typically is reacted with polymers including polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment.
  • PEG polyethylene glycol
  • the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer).
  • polyethylene glycol is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (C1 -C10) alkoxy- or aryloxy-polyethylene glycol or polyethylene glycol-maleimide.
  • the antibody to be pegylated is an aglycosylated antibody. Methods for pegylating proteins are known in the art and can be applied to the antibodies of the invention.
  • the selective anti-tofacitinib antibodies having V H and V L sequences disclosed herein can be used to create new anti-tofacitinib antibodies by modifying the V H and/or V L sequences, or the constant region(s) attached thereto.
  • the structural features of an anti-tofacitinib antibody of the invention are used to create structurally related anti-tofacitinib antibodies that retain at least one functional property of the antibodies of the invention, such as binding to tofacitinib but not substantially binding to metabolite 1 and/or metabolite 2.
  • one or more CDR regions of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and/or 12D4.G6, or mutations thereof can be combined recombinantly with known framework regions and/or other CDRs to create additional, recombinantly-engineered, anti-tofacitinib antibodies, as discussed above.
  • Other types of modifications include those described in the previous section.
  • the starting material for the engineering method is one or more of the V H and/or V L sequences provided herein, or one or more CDR regions thereof.
  • the engineered antibody it is not necessary to actually prepare ⁇ i.e., express as a protein) an antibody having one or more of the V H and/or V L sequences provided herein, or one or more CDR regions thereof. Rather, the information contained in the sequence(s) is used as the starting material to create a "second generation" sequence(s) derived from the original sequence(s) and then the "second generation" sequence(s) is prepared and expressed as a protein.
  • Standard molecular biology techniques can be used to prepare and express the altered antibody sequence.
  • the antibody encoded by the altered antibody sequence(s) is one that retains one, some, or all of the functional properties of the anti-tofacitinib antibodies described herein, which functional properties include, but are not limited to:
  • mutations can be introduced randomly or selectively along all or part of an anti-tofacitinib antibody coding sequence and the resulting modified anti-tofacitinib antibodies can be screened for binding activity and/or other functional properties as described herein. Mutational methods are well-known in the art.
  • Monoclonal antibodies (mAbs) of the present invention can be produced by a variety of techniques, including conventional monoclonal antibody methodology.
  • Mouse, rat, rabbit, camelid and human monoclonal antibodies of the invention can also be prepared using phage display methods for screening libraries of immunoglobulin genes from those, or many other, species.
  • Such phage display methods for isolating antibodies are established in the art and encompass phage display libraries prepared from immunized animals or humans or naive phage display libraries where the original starting material is not derived from immunized animals.
  • Phage display is well known in the art and is described, for example, in U.S. Patent No. 5,223,409, WO 91/17271 , WO92/20791 ; and WO 92/15679.
  • the monoclonal antibodies of the invention can also be prepared by culturing a host cell comprising a nucleic acid encoding the monoclonal antibody under suitable conditions and recovering said antibody, or antigen binding portion thereof.
  • a host cell comprising a nucleic acid encoding the monoclonal antibody under suitable conditions and recovering said antibody, or antigen binding portion thereof.
  • Such host cell culturing methods are well-known in the art.
  • This disclosure further provides a method for assessing the presence or concentration of tofacitinib in a sample, the method comprising providing a sample suspected of containing tofacitinib; contacting the sample or sample extract with an antibody specific for tofacitinib under conditions suitable for binding of the antibody to tofacitinib to form an assay mixture; and detecting binding of the antibody to tofacitinib.
  • this disclosure provides a method of determining the concentration of tofacitinib in a sample.
  • the method comprises providing a labeled competitor comprising tofacitinib coupled to a detectable label; providing a selective anti- tofacitinib antibody that binds to tofacitinib but not to metabolites 1 and/or 2; combining the sample, the selective anti-tofacitinib antibody, and the labeled competitor, wherein the tofacitinib in the sample competes with the labeled competitor for binding to the selective anti-tofacitinib antibody; and determining the concentration of tofacitinib in the sample by measuring the amount of the labeled competitor not bound to the antibody by detection of the label.
  • the amount of labeled competitor is detected in a sample that does not contain tofacitinib and the amount of label detected in the sample is compared to the amount of label present where tofacitinib may be present. The difference between the amount of label detected in the absence of tofacitinib and the amount of label in the sample that is suspected of containing tofacitinib is measured.
  • this disclosure provides competitive immunoassay kits for determining the presence or absence of tofacitinib in a sample are provided.
  • Illustrative competitive immunoassay kits such as an enzyme linked immunoassay (ELISA)
  • ELISA enzyme linked immunoassay
  • an antibody capable of specifically binding tofacitinib and a tofacitinib compound conjugated to a detectable label, wherein the conjugated tofacitinib compound is configured to compete with the tofacitinib in the sample to bind with the antibody, and wherein the label provides a signal indicative of a concentration of tofacitinib in the sample when the tofacitinib in the sample is present in therapeutic drug monitoring concentrations.
  • ELISA enzyme linked immunoassay
  • this disclosure provides a competitive immunoassay for determining the presence of tofacitinib in a sample at a concentration of drug within the therapeutic range of tofacitinib.
  • a competitive immunoassay for determining the presence of tofacitinib in a sample at a concentration of drug within the therapeutic range of tofacitinib.
  • immunoassays that monitor therapeutic drug concentrations may provide sensitivity across a broader range of tofacitinib concentrations.
  • the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 0 to about 1500 ng/ml.
  • the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 5 to about 1215 ng/ml. In another aspect, the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 5 to about 500 ng/ml. In another aspect, the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 5 to about 405 ng/ml. In another aspect, the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 15 to about 1215 ng/ml.
  • the labeled competitor may be derived from tofacitinib using a linkage that is the same or similar to that of the immunogenic tofacitinib conjugate.
  • the antibodies, immunogenic tofacitinib conjugates, labeled competitors and/or other conjugates described herein are also suitable for use with any of a number of other homogeneous and heterogeneous immunoassays with a range of detection systems.
  • the examples presented herein are not intended to be limiting.
  • the present invention provides tofacitinib conjugates that are useful for the preparation of immunogens and conjugates for use in immunoassays for the detection of tofacitinib.
  • a tofacitinib analog according to the present invention to an immunogenic carrier material
  • polyclonal or monoclonal antibodies can be produced and isolated, which are useful reagents for immunoassays for the detection of tofacitinib.
  • Coupling can be accomplished by any chemical reaction that will bind the label or carrier.
  • Illustrative tofacitinib immunoassays employ anti-tofacitinib antibodies that can be either polyclonal or monoclonal.
  • the antibody preparation used is induced by an immunogen described herein is formulated in an aqueous solution such as buffer, and the like or provided in an adjuvant or similar composition. The induced antibodies can be tested to determine specificity for tofacitinib.
  • the antibodies and labeled competitors described herein are also suitable for use with any of a number of other homogeneous and heterogeneous immunoassays with a range of detection systems.
  • the invention provides a kit for determining the concentration of tofacitinib in a sample, the kit comprising at least one selective anti-tofacitinib antibody.
  • the kit further comprises a labeled competitor comprising tofacitinib coupled to a detectable label.
  • the labeled competitor competes with the tofacitinib in a sample for binding to the anti-tofacitinib antibody.
  • the selective anti- tofacitinib antibody is produced using an immunogenic tofacitinib conjugate comprising tofacitinib coupled to an immunogenic carrier.
  • the kit may also include an applicator and/or instructional material for the use of the kit.
  • the invention includes a kit for determining the concentration of tofacitinib in a sample wherein the kit comprises at least one selective tofacitinib antibody that binds tofacitinib but does not substantially bind at least one metabolite selected from the group consisting of tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III.
  • the kit comprises at least one selective tofacitinib antibody that binds tofacitinib but does not substantially bind tofacitinib metabolite 1 of Formula II and does not substantially bind tofacitinib metabolite 2 of Formula III.
  • the kit further comprises an applicator.
  • the kit comprises an instructional material for the use of the kit.
  • the kit can detect a concentration of tofacitinib ranging from 5 ng/ml to 1215 ng/ml. In another aspect, the kit can detect a concentration of tofacitinib ranging from 5 ng/ml to 405 ng/ml. In yet another aspect, the kit can detect a concentration of tofacitinib ranging from 15 ng/ml to 1215 ng/ml.
  • tofacitinib-BSA conjugate (Formula V) was purified by desalting according to standard desalting methods known to those of skill in the art. Conjugation of tofacitinib with BSA was confirmed by Matrix-Assisted Laser Desorption Ionization (MALDI).
  • MALDI Matrix-Assisted Laser Desorption Ionization
  • biotinylated tofacitinib derivative of Formula VII One illustrative example of a tofacitinib labeled competitor is a biotinylated tofacitinib derivative of Formula VII.
  • the biotinylated tofacitinib derivative of Formula VII was produced as follows.
  • Polyclonal and monoclonal antibodies were produced by using conventional techniques, essentially as described by Kohler and Milstein, Nature 256:495-497(1975).
  • Three female Balb/C mice were immunized with 100 ⁇ g of the immunogenic tofacitinib-KLH conjugate of example 3 in complete Freund's adjuvant, administered by intraperitoneal (ip) injection.
  • Three subsequent injections were administered ip comprising 50 ⁇ g mouse of the immunogenic tofacitinib-KLH conjugate in incomplete Freund's adjuvant on days 10, 20, and 30 after initial injection. Serum was collected on day 37 and the presence of antibodies reactive to the antigen was determined by solid phase ELISA, as described below, against immunogenic tofacitinib-BSA conjugate of example 2.
  • Solid Phase ELISA The immunogenic tofacitinib-BSA conjugate of example 2 was immobilized to wells of a microtiter plate. Specifically, microtiter plates were coated with 2- 10 ⁇ g ml tofacitinib-BSA conjugate in a coating buffer (0.2 M carbonate buffer (BuPH carbonate-bicarbonate buffer pack, Pierce item #28382) for 1-2 hours at room temperature or overnight at 4°C, then saturated with a blocking buffer (PBS containing 1 % (w/v) BSA) for 1 hour at room temperature or overnight at 4°C, and washed 3x with washing buffer (PBS containing 0.5% (v/v) Tween 20).
  • a coating buffer 0.2 M carbonate buffer (BuPH carbonate-bicarbonate buffer pack, Pierce item #28382)
  • PBS containing 1 % (w/v) BSA a blocking buffer
  • washing buffer PBS containing 0.5% (v/v) Tween 20
  • the antibody samples i.e., the mouse serum or the hybridoma supernatants
  • a diluent 0.1 % solution of BSA in PBS.
  • the diluted antibody samples were added to microtiter plate and the plate was incubated for 1-2 hours at room temperature. After having washed away any unbound substances with washing buffer, the level of bound anti-tofacitinib antibody was determined using rabbit or goat anti-mouse peroxidase-conjugated secondary antibody (IgG specific) at recommended or experimentally derived dilution (usually 1/1000-1/10,000) in the diluent.
  • IgG specific rabbit or goat anti-mouse peroxidase-conjugated secondary antibody
  • FIG. 1 is a graph depicting binding of serum antibodies to the tofacitinib-BSA conjugate of example 2 as determined by solid phase ELISA. As demonstrated in Figure 1 , antibodies reactive to the antigen were present in each of mouse 1 , 2 and 3. As mouse 3 had the highest titer (defined as the dilution of serum at 50% maximum signal), this mouse was selected for hybridoma generation.
  • OPD o-phenylenediamine
  • Hybridoma generation The mouse demonstrating the highest serum antibody titer (defined as the dilution of serum at 50% maximum signal), mouse 3, received booster injection comprising 25 ⁇ g of antigen. Three days later, the mouse was sacrificed and its spleen cells were isolated and fused with NS1 myeloma cells following a standard fusion protocol. These mixtures of clones, called parental clones, were screened at 10 days after the fusion by solid-phase ELISA (described above) against immunogenic tofacitinib-BSA to identify parental clones that secreted antibodies capable of binding tofacitinib. Positive parental clones were selected for further analysis.
  • positive parental clones originating from the fusion were expanded from the 96 well plates to 24 well plates and were rescreened 3 days later to ensure that the clones were still producing antibody. From the rescreen, selected wells with positive hybridomas, (i.e., hybridomas that secreted antibodies capable of binding the immunogenic tofacitinib-BSA conjugate and that did not bind to metabolite-1-BSA, metabolite-2-BSA) were frozen for future use and sub-cloned to isolate positive cell lines.
  • positive hybridomas i.e., hybridomas that secreted antibodies capable of binding the immunogenic tofacitinib-BSA conjugate and that did not bind to metabolite-1-BSA, metabolite-2-BSA
  • the selected positive parent clones that secreted antibodies capable of binding immunogenic tofacitinib-BSA conjugate were subcloned by limiting dilution to obtain monoclonal hybridoma cell lines. Approximately 10 days after subcloning, small volumes of media were removed from the wells and screened by solid-phase ELISA to identify antibody- producing clones. Selected positive clones were expanded and rescreened to ensure that the clones were still producing antibody and confirm specificity. Up to two positive clones per parental line, including clones 6D9.A5 and 12D4.G6, were expanded for freezing 2 vials each. 1 ml supernatant was also collected for testing.
  • Example 5 In addition to the direct solid-phase ELISA described above in Example 5, alternative immunoassay formats were developed, including competitive a competitive immunoassay format for the detection of tofacitinib in experimental samples. Exemplary competitive immunoassay protocols are provided below.
  • Example 5 The direct solid-phase ELISA described above in Example 5 was converted to a competitive ELISA wherein a defined amount of monoclonal antibody replaced immune serum samples or hybridoma supernatants, a competitor (i.e., tofacitinib, tofacitinib metabolites) was then added to the monoclonal antibody solution, and binding of the monoclonal antibody to the tofacitinib-BSA conjugate in the presence and absence of the competitor was measured.
  • a competitor i.e., tofacitinib, tofacitinib metabolites
  • Microtiter plates were coated with 100 ⁇ /well 1 ⁇ g ml purified anti-tofacitinib antibody in a coating buffer for 3 hours at room temperature, then replaced with 300 ⁇ /well blocking buffer (PBS containing 1 % non-fat dry milk powder) for 30 minutes at room temperature, and washed 3x with washing buffer (PBS containing 0.5%) Tween 20).
  • TMB (3,3',5,5'-Tetramethylbenzidine) substrate was added, then color developed for 30 minutes, and the reaction was stopped by adding 50 ⁇ 2 N sulfuric acid. Absorbance at 450 nm was measured.
  • Binding of Anti-tofacitinib Monoclonal Antibodies to Biotinylated tofacitinib Selected antibodies were further tested for binding to tofacitinib by standard ELISA. Briefly, a total of 34 clones were screened for binding to biotinylated tofacitinib.
  • the format of the ELISA was as follows. Biotinylated tofacitinib (1 ng/mL) was incubated with antibody dilutions ranging from 1 :100 to 1 :1 ,638,400 on a pre-coated goat anti-mouse IgG plate for one hour.
  • FIG. 2 is a graph depicting binding of selected anti-tofacitinib antibodies to biotinylated tofacitinib as determined by ELISA. Eleven antibodies were found to bind to biotinylated tofacitinib in a concentration dependent manner. Data for ten of these antibodies are shown in Figure 2 and the data for antibody 8B5.F2 is not shown.
  • Antibodies were be further characterized as binding to tofacitinib or its metabolites by measuring cross reactivity using competitive immunoassay-3 mentioned above wherein tofacitinib metabolites as competitors.
  • eleven anti-tofacitinib monoclonal antibodies were screened for cross reactivity with metabolite 1 using competitive immunoassay.
  • 200 ⁇ /well of mixture containing anti-tofacitinib MAb(1/100 to 1/1638400 dilution), biotinylated tofacitinib (1 ng/ml), and Metabolite 1 (400 - 0.097 ng/ml) was added into goat anti-mouse IgG pre-coated plates, and incubated for 1 -2 hour at room temperature. After washing the plates, 200 ⁇ of streptavidin-HRP (50 ng/ml) was added and then incubated for 0.5-1 hour at room temperature. After washing the plates again, 200 ⁇ of TMB substrate was added, then developed for 30 minutes, and stopped by adding 50 ⁇ 2 N sulfuric acid. Absorbance at 450 nm was measured.
  • Figure 3 is a graph depicting cross reactivity of selected anti-tofacitinib antibodies to metabolite 1 as determined using the competitive immunoassay.
  • four antibodies demonstrated cross reactivity to metabolite 1 in a concentration dependent manner (e.g., clones 7C10.C1 1 , 1 B2.B9, 1 1 G10.F12, and 8B5.F2).
  • Seven clones were selective in that they bound tofacitinib but did not detectably bind metabolite 1 , e.g., 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5 (or 6D9.A8), 12H4.G2, 16F10.E6, and 12D4.G6.
  • These plots for the seven clones are shown as the lines going approximately straight across the top of the graph depicted in Figure 3. More specifically, the data used to prepare the graph shown in Figure 3 are as follows:
  • CDR3 ARIYYGIF SEQ ID NO:39
  • GQGYSYPYT SEQ ID NO:24
  • CDRs Complementarity Determining Regions
  • a competitive immunoassay format was used to determine whether tofacitinib inhibited the ability of the selected antibody clones that did not cross react with Metabolite- 1 to bind biotinylated tofacitinib.
  • the selected antibody clones that did not cross react with Metabolite-1 were assayed at their EC50 dilution, determined from the initial antibody titration (see Figure 2).
  • Table 4 summarizes the cross reactivity of clones 6D9.A8 and 12D4.G6 to Metabolite-1 and Metabolite-2.
  • Figure 4 is a graph depicting optimal dilution ranges of select monoclonal antibodies of the invention, e.g., 6D9.A8 (open circles) and 12D4.G6 (open squares) as determined by ELISA.
  • Table 5 summarizes the assay conditions and results of the ELISA.
  • Table 6 shows the data values used to generate the graph shown in Figure 5.
  • clone 12D4.G6 showed standard range of 5 to 405 ng/ml ( Figure 4), demonstrating that this monoclonal antibody is capable of detecting tofacitinib at levels as low as 5 ng/ml.
  • Figure 4 also showed standard range of 6D9.A8 was 15 - 1215 ng/ml.
  • tofacitinib was spiked into commercially available heparin human plasma to determine the effect of dilution on linearity and recovery of tofacitinib in human plasma sample.
  • Tofacitinib was spiked into Heparin-plasma (human) at a concentration of 4864 ng/ml and serially diluted 2-fold to test assay tolerance to plasma. Percent dilution linearity and percent recovery was calculated via an ELISA assay as described in Example 5.
  • tofacitinib dose response curves (0.001 to 1000 ng/ml) were run in the assay using antibody clones 6D9.A5, 6D9.A8, or 12D4.G6. The results, are shown in the Table 8 and Figure 5. Further, the data used to generate the graph shown in Figure 5 are set forth below in Table 9. The data disclosed herein indicate that 6D9.A5 has same standard range (15-1215 ng/ml) as 6D9.A8 and is capable of detecting tofacitinib in a sample at concentrations as low as about 15 ng/ml and as high as about 1215 ng/ml.
  • the replacement subclone antibody 6D9.A5 (replacing 6D9.A8) demonstrated a similar dose response activity to the parent drug as compared to subclone antibody 6D9.A8.
  • the data disclosed herein demonstrate that 6D9.A5 and 6D9.A8 antibodies are similarly more potent binders of the parent drug compared with antibody 12D4.G6.
  • 6D9.A5 antibody was further characterized by measuring cross reactivity to the metabolites using competitive immunoassay-3, wherein tofacitinib metabolites were used as competitors or using direct ELISA assay mentioned in Example 5, wherein the microtiter plates were coated with tofacitinib-BSA conjugate.
  • the data shown in Table 10 demonstrate that clone 6D9.A5 had similar cross reactivity to the metabolites as observed with clone 6D9.A8 and clone 12D4.G6. That is, like 12D4.G6, 6D9.A5 did not substantially bind metabolite 1 or 2.

Abstract

The invention provides selective tofacitinib antibodies, immunogenic tofacitinib conjugates that are useful as immunogenic molecules for the generation of antibodies specific for tofacitinib, along with methods for measuring the concentration of tofacitinib in a sample, processes for making the antibodies, and assays and kits for using the antibodies.

Description

ANTI-TOFACITINIB ANTIBODIES AND USES THEREOF FOR DRUG MONITORING
FIELD OF THE INVENTION
This invention relates to antibodies specific to tofacitinib that selectively bind to tofacitinib over two known metabolites. The selectivity of the anti-tofacitinib antibodies makes them particularly useful for immunoassays. The invention further relates to immunoassays or kits that include antibodies specific for tofacitinib.
BACKGROUND
This invention relates to antibodies specific to tofacitinib, which are useful, e.g., in assay methods and assay kits for monitoring blood levels of the drug.
Tofacitinib is a potent immunosuppressant in development for the treatment of rheumatoid arthritis (RA), psoriasis, inflammatory bowel disease, and other immunological diseases, as well as for the prevention of organ transplant rejection. Tofacitinib specifically inhibits Janus activated kinase 3 (JAK3), which has a pivotal role in cytokine signal transduction governing lymphocyte survival, proliferation, differentiation and apoptosis.
In certain circumstances, when organs such as kidney, heart, lung, bone marrow, and liver are transplanted in humans, the body will sometimes reject the transplanted tissue. One treatment of organ transplant rejection comprises suppression of the immune system in a controlled manner with immunosuppressant drugs such as tofacitinib. Immunosuppressant drugs are carefully administered to transplant recipients to prevent rejection of the foreign (i.e. non-self) tissue. Successful treatment with immunosuppressant drugs requires the measurement of drug concentrations, followed by subsequent dosage adjustments to maximize efficacy while minimizing toxicity. Monitoring blood levels of tofacitinib in patients treated with tofacitinib is thus very desirable in order to regulate the dosage. The appropriate dosage will maintain the minimum drug activity level sufficient for pharmacologic activity while avoiding any undue risk of side effects. Thus, need exists for a sensitive and reliable assay for measuring tofacitinib levels in patients that can be performed quickly and easily in a clinical setting as part of the development of tofacitinib as a pharmaceutical.
To support the use of tofacitinib in the clinical setting, particularly for use in the prevention of organ transplant rejection, a long-felt need exists for monitoring plasma levels of tofacitinib in many patients to ensure that therapeutic levels are maintained, as well as to guide adjustment of dosages in a timely fashion as needed. This activity is often referred to as Therapeutic Drug Monitoring (TDM). TDM plays a key role in helping clinicians maintain blood and plasma levels of immunosuppressive drugs within their respective therapeutic ranges. Variation in concentrations outside of the narrow therapeutic ranges may result in adverse clinical outcomes. TDM ensures that concentrations of drug are not too high or too l low, thereby reducing the risks of toxicity or rejection, respectively. Therapeutic monitoring of immunosuppressive drugs has generally been based on several choices of assay and biologic fluid (i.e., whole blood, plasma) appropriate for a particular drug.
A reliable liquid chromatography-mass spectrometry (LC-MS/MS) based assay is available for monitoring therapeutic levels of tofacitinib. Unfortunately, most clinical sites around the country and around the world are not set up to routinely perform LC-MS/MS assays in house. Clinical sites typically prefer relatively rapid, inexpensive, and easy to perform assays to streamline their TDM efforts. Accordingly, it would be advantageous to have immunoassays configured to detect tofacitinib in a patient's blood, serum, plasma, and/or other biological fluids or samples. Additionally, it would be advantageous to have tofacitinib-based immunogens for use in producing anti-tofacitinib antibodies.
Further, it would be advantageous to have specific anti-tofacitinib antibodies that bind tofacitinib but do not substantially bind at least one metabolite of tofacitinib. Such antibodies would be useful, among other things, to detect clinically relevant concentrations of tofacitinib in a sample from a patient undergoing therapy wherein tofacitinib is administered to the patient.
There have been no previous reports of monoclonal antibodies which recognize tofacitinib. There are inherent difficulties in making monoclonal antibodies to tofacitinib because tofacitinib is not immunogenic and is itself immunosuppressive. Moreover, as the metabolites of tofacitinib have not been well characterized in the literature, it is difficult to identify a monoclonal antibody capable of differentiating between tofacitinib and its metabolites. A need exists for an accurate assay to detect active tofacitinib but not its inactive metabolites. The present invention meets that need.
BRIEF SUMMARY OF THE INVENTION
The present invention is directed to novel immunogenic tofacitinib conjugates, as well as labeled tofacitinib competitors. The present invention is also directed to polyclonal and monoclonal antibodies generated using the immunogenic tofacitinib conjugates. These antibodies, conjugates, and competitors are useful in, among other things, immunoassays for the detection of tofacitinib in a sample.
The present invention includes an immunogenic tofacitinib conjugate comprising tofacitinib coupled to an immunogenic carrier, wherein the immunogenic carrier is a protein selected from the group consisting of keyhole limpet hemocyanin and bovine serum albumin, and further wherein
(a) the immunogenic tofacitinib conjugate has the structure
Figure imgf000005_0001
N-BSA
Formula V
wherein BSA is bovine serum albumin; or
(b) the immunogenic tofacitinib conjugate has the structure
Figure imgf000005_0002
N-KLH
Formula VI
wherein KLH is keyhole limpet hemocyanin.
The invention includes a method for assessing the concentration of tofacitinib in a sample. The method comprises:
(a) providing a sample suspected of containing tofacitinib;
(b) contacting the sample or sample extract with an antibody specific for tofacitinib under conditions suitable for binding of the antibody to tofacitinib to form an assay mixture; and
(c) detecting binding of the antibody to tofacitinib.
The invention includes an isolated antibody having greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
Figure imgf000006_0001
Formula II Formula III.
In one aspect, the isolated antibody has a binding affinity for tofacitinib that is 25, 30, 40, or 50 times the binding affinity for metabolite 1 or metabolite 2.
In another aspect, the antibody is obtained using an immunogenic tofacitinib conjugate compound comprising tofacitinib coupled to an immunogenic carrier.
The invention includes a method of determining the amount of tofacitinib in a sample. The method comprises:
providing known amount of a labeled competitor comprising tofacitinib coupled to a detectable label;
providing a selective anti-tofacitinib antibody;
combining the sample, the selective anti-tofacitinib antibody and the labeled competitor, wherein the tofacitinib in the sample competes with the labeled competitor for binding to the selective anti-tofacitinib antibody; and
determining the amount of tofacitinib in the sample by measuring the amount of labeled competitor not bound to antibody by detection of the label.
In one aspect, the selective anti-tofacitinib antibody has a greater binding affinity for t bolite 1 of Formula II
Figure imgf000006_0002
Formula II citinib metabolite 2 of Formula III
Figure imgf000007_0001
Formula III.
In one aspect, the antibody has greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
Figure imgf000007_0002
Formula II Formula III,
The invention includes a kit for determining the amount of tofacitinib in a sample. The kit comprises:
a labeled competitor comprising tofacitinib coupled to a detectable label; and at least one selective anti-tofacitinib antibody; wherein the labeled competitor competes with the tofacitinib in a sample for binding to the anti-tofacitinib antibody.
In one aspect, the selective anti-tofacitinib antibody has a greater binding affinity for tofacitinib than for at least one tofacitinib metabolite selected from the group consisting of tofacitinib metabolite 1 of Formula II
Figure imgf000007_0003
Formula II Formula III
Figure imgf000008_0001
Formula III.
In another aspect, the antibody has greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
Figure imgf000008_0002
Formula II Formula III.
In another aspect, the kit can be used to detect tofacitinib in a sample at a concentration ranging from about 5 ng/ml to about 1215 ng/ml and comprises
(a) an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24; and/or
(b) an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
The invention includes a competitive immunoassay kit for determining the concentration of tofacitinib in a sample. The competitive immunoassay comprises: at least one selective anti-tofacitinib antibody; a tofacitinib compound conjugated to a detectable label; wherein the conjugated tofacitinib compound competes with the tofacitinib in the sample to bind with the antibody; and wherein the label provides a signal indicative of the concentration of tofacitinib in the sample when the tofacitinib in the sample is present in a therapeutic drug monitoring concentration.
In one aspect, the antibody has greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
Figure imgf000009_0001
Formula II Formula III.
The invention includes an isolated antibody that binds to tofacitinib but does not substantially bind to at least one tofacitinib metabolite selected from the group consisting of a tofacitinib metabolite 1 of Formula II and a tofacitinib metabolite 2 of Formula III
Figure imgf000009_0002
Formula II Formula III.
In one aspect, the isolated antibody does not substantially bind a tofacitinib metabolite 1 of Formula II and a tofacitinib metabolite 2 of Formula I II
Figure imgf000009_0003
Formula II Formula III.
In another aspect, the antibody can detect tofacitinib in a sample but not substantially detect a tofacitinib metabolite in the sample, and wherein the amount of tofacitinib ranges from about 5 ng/mL to 1215 ng/mL, from about 5 ng/mL to 405 ng/mL, or about 15 ng/mL to 1215 ng/mL.
The invention includes an isolated antibody having greater binding affinity for tofacitinib than for at least one tofacitinib metabolite selected from the group consisting of a tofacitinib metabolite 1 of Formula II and a tofacitinib metabolite 2 of Formula III. The antibody comprises a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain comprises three complementarity determining regions (HCDR1 , HCDR2, and HCDR3) selected from the group consisting of:
(a) a HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:13, SEQ ID NO:19, SEQ ID NO:30, and SEQ ID NO:37;
(b) a HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:14, SEQ ID NO:20, SEQ ID NO:25, SEQ ID NO:31 , and SEQ ID NO:38;
(c) a HCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:15, SEQ ID NO:21 , SEQ ID NO:26, SEQ ID NO:32, and SEQ ID NO:39; and wherein the light chain variable domain comprises three CDRs (LCDR1 , LCDR2, and LCDR3) selected from the group consisting of:
(d) a LCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:16, SEQ ID NO:22, SEQ ID NO:27, SEQ ID NO:33, and SEQ ID NO:34;
(e) a LCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:17, SEQ ID NO:23, SEQ ID NO:28, and SEQ ID NO:35; and
(f) a LCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:18, SEQ ID NO:24, SEQ ID NO:29, and SEQ ID NO:34.
In one aspect, the antibody comprises
(a) a HCDR1 comprising the amino acid sequence of SEQ ID NO:13, a HCDR2 comprising the amino acid sequence of SEQ ID NO:14, a HCDR3 comprising the amino acid sequence of SEQ ID NO:15, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:16, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18;
(b) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(c) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:25, a HCDR3 comprising the amino acid sequence of SEQ ID NO:26, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:27, a LCDR2 comprising the amino acid sequence of SEQ ID NO:28, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:29;
(d) a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(e) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:33, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18;
(f) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:34, a LCDR2 comprising the amino acid sequence of SEQ ID NO:35, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:36;
(g) a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(h) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:10 and SEQ ID NO:12, and further comprising a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:9, and SEQ ID NO:1 1 ;
(i) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:1 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:2;
(j) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:3 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(k) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4; (I) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(m) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:5 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:6;
(n) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:8 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:9;
(o) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:10 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:1 1 ;
(p) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID N03, SEQ ID NO:7 and SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4; and
(q) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:8, SEQ ID NO:10, and SEQ ID NO:12 and a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:4, SEQ ID NO:9 and SEQ ID NO:1 1.
In another aspect, the antibody is selected from the group consisting of:
(a) an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24:
(b) an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(c) an antibody comprising a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:7 and SEQ ID NO:12 and further comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4; (d) an antibody comprising a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and further comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4; and
(e) an antibody comprising a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:12 and further comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4.
In one aspect, the antibody is capable of detecting tofacitinib in a sample at a concentration ranging from about 15 ng/ml to 1215 ng/ml.
In another aspect, the antibody is capable of detecting tofacitinib in a sample at a concentration ranging from about 5 ng/ml to 405 ng/ml.
In yet another aspect,
The invention includes a nucleic acid encoding an antibody comprising
(a) a HCDR1 comprising the amino acid sequence of SEQ ID NO:13, a HCDR2 comprising the amino acid sequence of SEQ ID NO:14, a HCDR3 comprising the amino acid sequence of SEQ ID NO: 15, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO: 16, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18;
(b) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(c) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:25, a HCDR3 comprising the amino acid sequence of SEQ ID NO:26, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:27, a LCDR2 comprising the amino acid sequence of SEQ ID NO:28, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:29;
(d) a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(e) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:33, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18; (f) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:34, a LCDR2 comprising the amino acid sequence of SEQ ID NO:35, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:36;
(g) a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(h) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1 , SEQ ID NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:10 and SEQ ID NO:12, and further comprising a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:9, and SEQ ID NO:1 1 ;
(i) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:1 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:2;
(j) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:3 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(k) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(I) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(m) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:5 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:6;
(n) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:8 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:9;
(o) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:10 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:1 1 ; (p) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID N03, SEQ ID NO:7 and SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4; and
(q) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:8, SEQ ID NO:10, and SEQ ID NO:12 and a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:4, SEQ ID NO:9 and SEQ ID NO:1 1.
In one aspect, the invention includes a host cell comprising the nucleic acid.
In another aspect, the invention includes a method of producing the antibody. The method comprises culturing the host cell under conditions wherein the antibody is produced. In a further aspect, the method comprises isolating the antibody.
BRIEF DESCRIPTION OF THE DRAWINGS
The disclosed subject matter is particularly pointed out and distinctly claimed in the claims at the conclusion of the specification. The foregoing and other objects, features, and advantages of the disclosed embodiments are apparent from the following detailed description taken in conjunction with the accompanying drawings in which:
Figure 1 is a graph depicting binding of serum antibodies to the tofacitinib-BSA conjugate of example 2 as determined by solid phase ELISA.
Figure 2 is a graph depicting binding of selected anti-tofacitinib antibodies to biotinylated tofacitinib as determined by ELISA.
Figure 3 is a graph depicting cross reactivity of selected anti-tofacitinib antibodies to metabolite 1 as determined by ELISA.
Figure 4 is a graph depicting optimal dilution ranges of select monoclonal antibodies of the invention as determined by ELISA.
Figure 5 is a graph depicting optimal dilution ranges of select monoclonal antibodies of the invention as determined by ELISA.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides novel immunogenic tofacitinib conjugates comprising tofacitinib coupled to an immunogenic carrier, as well as labeled tofacitinib competitors. The present invention is also directed to polyclonal and monoclonal antibodies generated using the immunogenic tofacitinib conjugates.
The present invention also provides polyclonal and monoclonal antibodies which are specific for tofacitinib. In some embodiments, the antibodies of the invention are produced in response to inoculation with a novel immunogenic tofacitinib conjugate comprising tofacitinib linked to an immunogenic carrier. These antibodies, conjugates, and competitors are useful in immunoassays for the detection of tofacitinib in biological fluids. Assay kits comprising these antibodies, conjugates and competitors are well suited for use in a clinical setting and provide far more accurate and reproducible results than was previously possible. The antibodies are also useful in the purification and isolation of tofacitinib.
The term "antibody" as referred to herein includes whole antibodies and any antigen binding fragment (i.e., "antigen-binding portion") or single chains thereof. An "antibody" refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding portion thereof. An "antibody" also refers to an IgA, IgD, IgE, IgG, IgM antibody subtype. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region (CH). The heavy chain constant region is typically comprised of three domains, Cm, CH2 and CH3- Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors.
A "CDR" of a variable domain are amino acid residues within the variable region that are identified in accordance with the definitions of the Kabat, Chothia, the accumulation of both Kabat and Chothia, AbM, contact, and/or conformational definitions or any method of CDR determination well known in the art. Antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. See, e.g., Kabat et al., 1992,
Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, NIH, Washington D.C. The positions of the CDRs may also be identified as the structural loop structures originally described by Chothia and others. See, e.g., Chothia et al., 1989, Nature 342:877-883. Other approaches to CDR identification include the "IMGT definition" (Lefranc, M.-P. et al., 1999, Nucleic Acids Res. 27:209-212) and the "AbM definition," which is a compromise between Kabat and Chothia and is derived using Oxford Molecular's AbM antibody modeling software (now Accelrys®), or the "contact definition" of CDRs based on observed antigen contacts, set forth in MacCallum et al., 1996, J. Mol. Biol. 262:732-745. In another approach, referred to herein as the "conformational definition" of CDRs, the positions of the CDRs may be identified as the residues that make enthalpic contributions to antigen binding. See, e.g., Makabe et al., 2008, J. Biol. Chem. 283:1 156-1 166. Still other CDR boundary definitions may not strictly follow one of the above approaches, but will nonetheless overlap with at least a portion of the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. As used herein, a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches. The methods used herein may utilize CDRs defined according to any of these approaches. For any given embodiment containing more than one CDR, the CDRs may be defined in accordance with any of Kabat, Chothia, extended, AbM, contact, and/or conformational definitions.
The term "antigen-binding portion" or "antigen-binding fragment" of an antibody (or simply "antibody portion"), as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., tofacitinib). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full- length antibody. Examples of binding fragments encompassed within the term "antigen- binding portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the V|_, VH, C|_ and Cm domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and Cm domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., 1989, Nature 341 :544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al., 1988, Science 242:423-426; and Huston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen- binding portion" of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
An "isolated antibody", as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that specifically binds tofacitinib is substantially free of antibodies that specifically bind antigens other than tofacitinib). Moreover, an isolated antibody may be substantially free of other cellular material and/or chemicals.
The terms "monoclonal antibody" or "monoclonal antibody composition" as used herein refer to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
The term "recombinant antibody", as used herein, includes all antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from a host cell transformed to express the antibody, (b) antibodies isolated from a recombinant, combinatorial antibody library, and (c) antibodies prepared, expressed, created or isolated by any other means that involve splicing of immunoglobulin gene sequences to other DNA sequences. Such recombinant antibodies comprise variable regions in which the framework and CDR regions are derived from mouse germline immunoglobulin sequences. In certain embodiments, however, such recombinant mouse antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to mouse germline VH and VL sequences, may not naturally exist within the mouse antibody germline repertoire in vivo.
As used herein, "isotype" refers to the antibody class (e.g., IgM or IgG) that is encoded by the heavy chain constant region genes.
The phrases "an antibody recognizing an antigen" and "an antibody specific for an antigen" are used interchangeably herein with the term "an antibody which binds specifically to an antigen."
The term "antibody derivatives" refers to any modified form of the antibody, e.g., a conjugate of the antibody and another agent or antibody.
The term "humanized antibody" is intended to refer to antibodies in which CDR sequences derived from the germline of a non-human species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications may be made within the human framework sequences.
The term "chimeric antibody" is intended to refer to antibodies in which the variable region sequences are derived from one species and the constant region sequences are derived from another species, such as an antibody in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody. Chimeric antibody can also include an antibody where the V domain and C domain are each derived two different sources even if both are from the same species.
As used herein, an antibody that "specifically binds to tofacitinib" or "a selective tofacitinib antibody" refers to an antibody that binds to tofacitinib but which does not substantially bind a metabolite of tofacitinib. An antibody specifically binds tofacitinib where it does not detectibly binds to a tofacitinib metabolite 1 of Formula II and/or to a tofacitinib metabolite 2 of Formula III or binds to such metabolite to a much lesser extent. For instance, the metabolite does not substantially compete with tofacitinib for binding to the antibody as measured by, for example, a competitive binding assay. A selective tofacitinib antibody binds to tofacitinib with an affinity that is 5, 10, 15, 25, 30, 40, or 50 times greater than its affinity to metabolite 1 of Formula II or metabolite 2 of Formula III.
As used herein, the term "subject" includes any human or nonhuman animal. The term "nonhuman animal" includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows chickens, amphibians, reptiles, etc.
Various aspects of the invention are described in further detail in the following subsections.
A "variable region" of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination. As known in the art, the variable regions of the heavy and light chain each consist of four framework regions (FRs) connected by three complementarity determining regions (CDRs) also known as hypervariable regions, contribute to the formation of the antigen binding site of antibodies. If variants of a subject variable region are desired, particularly with substitution in amino acid residues outside of a CDR region (i.e., in the framework region), appropriate amino acid substitution, preferably, conservative amino acid substitution, can be identified by comparing the subject variable region to the variable regions of other antibodies which contain CDR1 and CDR2 sequences in the same canonical class as the subject variable region (Chothia and Lesk, J Mol Biol 196(4): 901-917, 1987). When choosing FR to flank subject CDRs, e.g., when humanizing or optimizing an antibody, FRs from antibodies which contain CDR1 and CDR2 sequences in the same canonical class are preferred.
A "CDR" of a variable domain are amino acid residues within the variable region that are identified in accordance with the definitions of the Kabat, Chothia, the accumulation of both Kabat and Chothia, AbM, contact, and/or conformational definitions or any method of CDR determination well known in the art. Antibody CDRs may be identified as the hypervariable regions originally defined by Kabat et al. See, e.g., Kabat et al., 1992, Sequences of Proteins of Immunological Interest, 5th ed., Public Health Service, NIH, Washington D.C. The positions of the CDRs may also be identified as the structural loop structures originally described by Chothia and others. See, e.g., Chothia et al., 1989, Nature 342:877-883. Other approaches to CDR identification include the "IMGT definition" (Lefranc, M.-P. et al., Nucleic Acids Res., 27, 209-212 (1999)) and the "AbM definition," which is a compromise between Kabat and Chothia and is derived using Oxford Molecular's AbM antibody modeling software (now Accelrys®), or the "contact definition" of CDRs based on observed antigen contacts, set forth in MacCallum et al., 1996, J. Mol. Biol., 262:732-745. In another approach, referred to herein as the "conformational definition" of CDRs, the positions of the CDRs may be identified as the residues that make enthalpic contributions to antigen binding. See, e.g., Makabe et al., 2008, Journal of Biological Chemistry, 283:1 156- 1 166. Still other CDR boundary definitions may not strictly follow one of the above approaches, but will nonetheless overlap with at least a portion of the Kabat CDRs, although they may be shortened or lengthened in light of prediction or experimental findings that particular residues or groups of residues or even entire CDRs do not significantly impact antigen binding. As used herein, a CDR may refer to CDRs defined by any approach known in the art, including combinations of approaches. The methods used herein may utilize CDRs defined according to any of these approaches. For any given embodiment containing more than one CDR, the CDRs may be defined in accordance with any of Kabat, Chothia, extended, AbM, contact, and/or conformational definitions.
The term "monoclonal antibody" (Mab) refers to an antibody that is derived from a single copy or clone, including e.g., any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced. Preferably, a monoclonal antibody of the invention exists in a homogeneous or substantially homogeneous population.
Antibodies of the invention can be produced using techniques well known in the art, e.g., recombinant technologies, phage display technologies, synthetic technologies or combinations of such technologies or other technologies readily known in the art (see, for example, Jayasena, S.D., 1999, Clin. Chem. 45:1628-1650 and Fellouse et al., 2007, J. Mol. Biol., 373(4):924-940).
The terms "specific binding," "selective binding" "selectively binds," or "specifically binds" as used herein refer to the ability to bind to a target compound or epitope with greater affinity than to a non-target compound. In an illustrative example, an antibody that specifically binds tofacitinib has a greater affinity for tofacitinib than for metabolites of tofacitinib, e.g., metabolite 1 of Formula II and metabolite 2 of Formula III. In some embodiments, "specifically binds" or "selectively binds" refers to binding to a target compound with an affinity that is at least 5, 10, 15, 20, 25, 30, 50, 100, 500, 1000 or more times greater than the affinity for a non-target.
The term "epitope" refers to that portion of a molecule capable of being recognized by and bound by an antibody at one or more of the antibody's antigen-binding regions. Epitopes often consist of a chemically active surface grouping of molecules such as amino acids or sugar side chains and have specific three-dimensional structural characteristics as well as specific charge characteristics. The term "antigenic epitope" as used herein, is defined as a portion of a polypeptide to which an antibody can specifically bind as determined by any method well known in the art, for example, by conventional immunoassays. A "nonlinear epitope" or "conformational epitope" comprises noncontiguous polypeptides (or amino acids) within the antigenic protein to which an antibody specific to the epitope binds. As used herein, the terms "immunogen" and "immunogenic" are meant to refer to substances capable of producing or generating an immune response in an organism. An immunogen can also be antigen. Usually, an immunogen has a fairly high molecular weight (e.g., greater than 10,000), thus, a variety of macromolecules such as proteins, lipoproteins, polysaccharides, some nucleic acids, and certain of the teichoic acids, can be an immunogen.
"Haptens" are partial or incomplete antigens. They are usually protein-free substances, mostly of low molecular weight, which are not generally capable of stimulating antibody formation, but which do react with antibodies. Antibodies to a hapten may be generated by coupling a hapten to a high molecular weight antigenic carrier (e.g., an immunogen) and then injecting this coupled product, i.e., an immunogenic conjugate, into a human or animal subject. For example, tofacitinib is a hapten.
A "carrier" or "immunogenic carrier," as the terms are used herein, is an immunogenic substance, commonly a protein, that can join with a hapten, thereby enabling the hapten to induce an immune response and elicit the production of antibodies that can bind specifically with the antigen (hapten). Carrier substances include proteins, glycoproteins, complex polysaccharides, particles, and nucleic acid that are recognized as foreign and thereby elicit an immunologic response from the host. Various proteins may be employed as a polypeptide immunogenic carrier. These proteins include albumins and serum proteins, e.g., globulins, ocular lens proteins, lipoproteins, etc. Illustrative proteins include bovine serum albumin (BSA), keyhole limpet hemocyanin (KLH), egg ovalbumin, bovine gamma-globulin (BGG), etc. Alternatively, synthetic polypeptides may be used. The immunogenic carrier can also be a polysaccharide such as, e.g., starches, glycogen, cellulose, carbohydrate gums such as gum arabic, agar, and so forth. The polysaccharide can also contain polypeptide residues and/or lipid residues. The immunogenic carrier can also be a polynucleotide either alone or conjugated to one of the polypeptides or polysaccharides mentioned above.
As used herein, the term "immunogenicity" is meant to refer to the ability of a molecule to induce an immune response, which may be determined both by the intrinsic chemical structure of the injected molecule and by whether or not the host animal's immune system recognizes the compound. Small changes in the structure of an antigen can greatly alter the immunogenicity of a compound, and have been used extensively as a general procedure to increase the chances of raising an antibody, particularly against well-conserved antigens. For example, these modification techniques either alter regions of the immunogen to provide better sites for T-cell binding or expose new epitopes for B-cell binding.
As used herein the term "label" refers to any molecule that produces, or can be induced to produce, a detectable signal. The label can be conjugated to an analyte, an immunogen, and/or an antibody. Non-limiting examples of labels include radioactive isotopes, enzymes, enzyme fragments, enzyme substrates, enzyme inhibitors, coenzymes, catalysts, fluorophores, dyes, chemiluminescers, luminescers, sensitizers, non-magnetic or magnetic particles, solid supports, liposomes, ligands, receptors, and hapten radioactive isotopes.
The term "labeled," with regard to a specific antibody or a labeled competitor, includes direct labeling by coupling (i.e., physically linking) a detectable substance to the antibody or labeled competitor, as well as indirect labeling of the antibody or labeled competitor by coupling it with another reagent that is, in turn, directly labeled. An example of indirect labeling includes detection of a primary antibody using a fluorescent-labeled secondary antibody. In vitro techniques for detection of an antigen of the invention include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitation, and immunofluorescence.
The antibodies, labeled competitors, and potential therapeutic compounds described herein are also suitable for use with any of a number of other homogeneous and heterogeneous immunoassays with a range of detection systems.
As used herein, the term "antigenic compound" refers to a compound used to produce an immune response. Illustratively, the antigenic compound is a hapten, for example tofacitinib, linked to an immunogenic carrier. The antigenic compound is used to generate the desired antibodies.
The term "labeled competitor" as used herein is a molecule capable of specific binding to antibodies having specificity for tofacitinib, wherein the molecule is linked to a detectable label or tracer. Illustratively, the molecule is tofacitinib or a derivative or analyte thereof.
The term "biological sample" includes, but is not limited to, any quantity of a substance isolated or derived from a living subject or formerly living subject. The term is intended to include tissues, cells, and biological fluids isolated from a subject, as well as tissues, cells, and fluids present within a subject. Subjects include, but are not limited to, chicken, humans, mice, monkeys, rats, rabbits, horses, camelids, and other animals. Such substance include, but are not limited to, blood, plasma, serum, semen, urine, tears, saliva, cells, organs, tissues, bone, bone marrow, lymph, lymph nodes, synovial tissue or fluid, chondrocytes, synovial macrophages, endothelial cells, and skin.
In one aspect, this disclosure provides derivatives of tofacitinib which are useful as immunogenic molecules for the generation of antibodies specific for tofacitinib for measuring levels of tofacitinib.
In another aspect, this disclosure provides methods and kits for selectively detecting tofacitinib in a sample. Tofacitinib (Formula I) is an orally available, potent selective inhibitor of JAK3 that is in development for the treatment of rheumatoid arthritis (RA) and other autoimmune disorders, such as inflammatory bowel disease, ankylosing spondylitis, psoriasis, psoriatic arthritis, and the prevention of transplant rejection.
Figure imgf000023_0001
Formula II Formula III
Implementing an immunoassay for the detection of a small molecule, such as tofacitinib, can be difficult. In some cases, small molecules lack antigenicity, making it difficult to generate antibodies against them. This challenge is compounded for tofacitinib because it has such potent immunosuppressive properties. To increase the immunogenicity of small molecules, larger immunogenic compounds, including but not limited to bovine serum albumin, ovalbumin, keyhole limpet hemocyanin, and the like, can be coupled to the drug. Detection of the drug in a sample generally requires the use of a detectable label conjugated to an antibody, tofacitinib, or tofacitinib analog.
Prior to the present invention, it was not expected that tofacitinib could be rendered more immunogenic by conjugation to an immunogenic carrier. Thus, surprisingly, in one aspect, this disclosure provides an immunogenic tofacitinib conjugate comprising tofacitinib coupled to an immunogenic carrier, wherein the immunogenic carrier is a protein, glycoprotein, complex polysaccharide, or nucleic acid that is recognized by the immunized animal as foreign, resulting in an immunologic response. In one illustrative example, the immunogenic carrier is a protein selected from the group consisting of keyhole limpet hemocyanin (KLH) and bovine serum albumin (BSA).
In one embodiment, this disclosure provides an immunogenic tofacitinib conjugate of formula V, having the structure
Figure imgf000024_0001
N-BSA
Formula V
wherein BSA is bovine serum albumin.
In another embodiment, this disclosure provides an immunogenic tofacitinib conjugate of formula VI, having the structure
Figure imgf000024_0002
N-KLH
Formula VI
wherein KLH is Keyhole Limpet Hemocyanin.
The immunogenic tofacitinib conjugates of the present invention can be used to elicit antibodies using known antibody producing and screening procedures. The immunogenic tofacitinib conjugates can be injected into appropriate animal hosts to stimulate the production of antibodies. The antibodies so produced can be harvested for direct use in immunoassays configured to detect tofacitinib. In some cases, it is desirable to have a monoclonal antibody that is specific to tofacitinib, and which selectively binds to tofacitinib over tofacitinib metabolites. In some embodiments, the antibody has a binding affinity for tofacitinib that is about 25, 30, 40, or 50 times the binding affinity for metabolite 1 or metabolite 2. In some embodiments, the antibody binds to metabolite 1 or metabolite 2 with an affinity that is less than about 5%, 4%, 3%, 2%, 1 %, or 0.5% of the binding affinity to tofacitinib. Selective Anti-Tofacitinib Antibodies
The antibodies of the present invention are characterized by the fact that they bind specifically to tofacitinib (Formula I), but which do not substantially bind to one or both known tofacitinib metabolites, "metabolite 1 of Formula II" and "metabolite 2 of Formula III," and methods of making and using these anti-tofacitinib antibodies. A selective tofacitinib antibody binds to tofacitinib with an affinity that is 5, 10, 15, 25, 30, 40, or 50 times greater than its affinity to metabolite 1 of Formula II or metabolite 2 of Formula III.
Standard assays to evaluate the binding ability of the antibodies toward tofacitinib and its metabolites are known in the art, including for example, ELISAs, Western blots, radioimmunoassays, and flow cytometry analysis. Suitable assays are described in detail in the Examples. The binding kinetics (e.g., binding affinity) of the antibodies also can be assessed by standard assays known in the art, such as by Biacore SPR analysis and Octet analysis.
The antibodies of the invention include mouse monoclonal antibodies 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6. The VH amino acid sequences of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in Table 2, and are set forth in SEQ ID NOs: 1 , 3, 5, 7, 8, 10 and 12, respectively. The VL amino acid sequences of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in Table 2 and are set forth in SEQ ID NOs: 2, 4, 6, 4, 9, 1 1 and 4, respectively.
The VH and VL sequences may be "mixed and matched" to create other selective tofacitinib binding molecules of the invention. Tofacitinib binding of such mixed and matched antibodies, as well as the binding of the antibody to one or both metabolites 1 and 2, can be tested using the binding assays described above and in the Examples. Optionally, when VH and VL chains are mixed and matched, a VH sequence from a particular VHA L pairing is replaced with a structurally similar VH sequence. Likewise, optionally a VL sequence from a particular VHA L pairing is replaced with a structurally similar VL sequence.
Accordingly, in one aspect, the invention provides an isolated monoclonal antibody, or antigen binding portion thereof, comprising:
(a) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1 , SEQ I D NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:10 and SEQ ID NO:12, and further comprising a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2, SEQ I D NO:4, SEQ ID NO:6, SEQ ID NO:9, and SEQ ID NO:1 1 ; (b) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:1 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:2;
(c) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:3 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(d) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(e) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(f) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:5 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:6;
(g) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:8 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:9;
(h) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:10 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:1 1 ;
(i) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID N03, SEQ ID NO:7 and SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4; and
(j) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ I D NO:8, SEQ ID NO:10, and SEQ ID NO:12 and a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:4, SEQ ID NO:9 and SEQ ID NO:1 1 .
In another aspect, the invention provides antibodies that comprise the heavy chain and/or light chain CDR1 s, CDR2s and CDR3s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6, or combinations thereof. The amino acid sequences of the VH CDR1 S of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs: 13, 19, 19, 30, 19, 19, and 37, respectively. The amino acid sequences of the VH CDR2s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs: 14, 20, 25, 31 , 20, 20, and 38, respectively. The amino acid sequences of the VH CDR3s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs:15, 21 , 26, 32, 21 , 21 , and 39, respectively. The amino acid sequences of the VL CDR1 s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs:16, 22, 27, 22, 33, 34, and 22, respectively. The amino acid sequences of the VL CDR2s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs:17, 23, 28, 23, 17, 35 and 23, respectively. The amino acid sequences of the VL CDR3s of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 are shown in SEQ ID NOs:18, 24, 29, 24, 18, 36, and 24, respectively. The CDR regions are delineated using the Kabat system (Kabat, E. A., et al. Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242 (1991 )).
The heavy and light chain CDRs for the antibody 5A3.E5 comprise SEQ ID NOs: 13, 14, 15, 16, 17, 18. The heavy and light chain CDRs for the antibody 10A6.C5 comprise SEQ ID NOs: 19, 20, 21 , 22, 23, and 24. The heavy and light chain CDRs for the antibody 10F10.H5 comprise SEQ ID NOs:19, 25, 26, 27, 28, and 29. The heavy and light chain CDRs for the antibody 6D9.A5 comprise SEQ ID NOs:30, 31 , 32, 22, 23, and 24. The heavy and light chain CDRs for the antibody 12H4.G2 comprise SEQ ID NOs:19, 20, 21 , 33, 17, and 18. The heavy and light chain CDRs for the antibody 16F10.E6 comprise SEQ ID NOs:19, 20, 21 , 34, 35, and 36. The heavy and light chain CDRs for the antibody 12D4.G6 comprise SEQ ID NOs:37, 38, 39, 22, 23, and 24.
In yet another embodiment, an antibody of the invention comprises heavy and light chain variable regions and/or heavy chain and light chain CDR1 s, CDR2s and CDR3s comprising amino acid sequences that are homologous to the amino acid sequences of the antibodies previously described herein, and wherein the antibodies retain the desired functional properties of the selective anti-tofacitinib antibodies of the invention.
In various embodiments, the antibody can be, for example, a mouse antibody, a humanized antibody or a chimeric antibody derived from a mouse anti-tofacitinib antibody. In other embodiments, the VH and/or VL amino acid sequences may be 85%, 90%, 95%, 96%, 97%, 98% or 99% homologous to the sequences set forth above. As used herein, the percent homology between two amino acid sequences is equivalent to the percent identity between the two sequences. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
In certain embodiments, an antibody of the invention comprises heavy and light chain variable regions and/or heavy chain and light chain CDR1 s, CDR2s and CDR3s, wherein one or more of these sequences comprise specified amino acid sequences based on the antibodies described herein, or conservative modifications thereof, and wherein the antibodies retain the desired functional properties of the anti-tofacitinib antibodies of the invention.
In alternative embodiments, the antibody can be, for example, mouse antibodies, humanized antibodies or chimeric antibodies.
As used herein, the term "conservative sequence modifications" is intended to refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody of the invention by standard techniques known in the art, such as site- directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, one or more amino acid residues within the CDR regions of an antibody of the invention can be replaced with other amino acid residues from the same side chain family and the altered antibody can be tested for retained function using the functional assays described herein.
In another embodiment, the invention provides an antibody that binds to the same epitope on tofacitinib as the anti-tofacitinib antibodies of the invention (i.e., an antibody that has the ability to compete for binding to tofacitinib with an antibody of the invention). In alternative embodiments, the reference antibody for competition studies can be the monoclonal antibody 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6. Such competing antibodies can be identified based on their ability to compete with 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, or 12D4.G6 for binding of tofacitinib in standard binding assays. For example, Biacore analysis, Octect analysis, ELISA assays or flow cytometry may be used to demonstrate cross-competition with the antibodies of the current invention. The ability of a test antibody to inhibit the binding of, for example, 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, or 12D4.G6, to tofacitinib, wherein the test antibody does not substantially bind metabolite 1 and/or metabolite 2, demonstrates that the test antibody can compete with 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 for binding to tofacitinib and thus may bind the same epitope on tofacitinib as 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6. In a further embodiment, the antibody that binds to the same epitope on tofacitinib as 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6 is a mouse monoclonal antibody. Such monoclonal antibodies can be prepared and isolated as described in the Examples or by a wide variety of methods well-known in the art. In other embodiments, the antibody that competes with the antibody is a human, humanized or mouse antibody.
An antibody of the invention can be prepared using an antibody comprising at least one of the VH and/or VL sequence disclosed herein as starting material to engineer a modified antibody, which may have properties that differ from the starting antibody but which may bind the same, or substantially the same, epitope as the starting material antibody. An antibody can be engineered by modifying one or more residues within one or both variable regions (i.e., VH and/or VL), for example within one or more CDR regions and/or within one or more framework regions. Additionally or alternatively, an antibody can be engineered by modifying residues within the constant region(s), for example to alter the effector function(s) of the antibody.
The present invention provides nucleic acids encoding the tofacitinib specific antibody of the invention. Nucleic acids encoding the antibodies of the invention can be generated by methods known in the art. Also, as would be understood by one skilled in the art, due to the degeneracy of the nucleic acid code, a wide variety of nucleic acid sequences can encode the amino acid sequence of the antibody of the invention.
One type of variable region engineering that can be performed is CDR grafting. Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain complementarity determining regions (CDRs). For this reason, the amino acid sequences within CDRs are more diverse between individual antibodies than sequences outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of specific naturally occurring antibodies by constructing expression vectors that include CDR sequences from the specific naturally occurring antibody grafted onto framework sequences from a different antibody with different properties (see, e.g., Riechmann et al., 1998, Nature 332:323-327; Jones et al., 1986, Nature 321 :522-525; Queen et al., 1989, Proc. Natl. Acad. See. U.S.A. 86:10029-10033; U.S. Patent Nos. 5,225,539; 5,530,101 ; 5,585,089; 5,693,762 and 6,180,370).
Accordingly, another embodiment of the invention pertains to an isolated monoclonal antibody, or antigen binding portion thereof, comprising a heavy chain variable region comprising CDR1 , CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 13, 19, 30, and 37; SEQ ID NOs:14, 20, 25, 31 , and 38; and SEQ ID NOs:15, 21 , 26, 32, and 39, respectively, and a light chain variable region comprising CDR1 , CDR2, and CDR3 sequences comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:16, 22, 27, 33, and 34; SEQ ID NOs: 17, 23, 28, and 35; and SEQ ID NOs:18, 24, 29, and 36, respectively. Such antibodies contain the VH and VL CDR sequences of monoclonal antibodies 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and 12D4.G6, which may contain a different framework sequence from the antibodies. Such framework sequences can be obtained from public DNA databases or published references that include germline antibody gene sequences.
Another type of variable region modification is to mutate amino acid residues within the VH and/or VL CDR1 , CDR2 and/or CDR3 regions to thereby improve one or more binding properties (e.g., affinity) of the antibody of interest. Site-directed mutagenesis or PCR- mediated mutagenesis can be performed to introduce the mutation(s) and the effect on antibody binding, or other functional property of interest, can be evaluated in in vitro or in vivo assays as described herein and provided in the Examples. Optionally, conservative modifications (as discussed above) are introduced. The mutations may be amino acid substitutions, additions or deletions. Moreover, typically no more than one, two, three, four or five residues within a CDR region are altered.
In yet another embodiment, the invention provides isolated anti-phospho-tau monoclonal antibodies, or antigen binding portions thereof, comprising a heavy chain variable region comprising:
(a) a VH CDR1 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs:13, 19, 30, and 37, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 13, 19, 30, and 37; (b) a VH CDR2 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 14, 20, 25, 31 , and 38, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 14, 20, 25, 31 , and 38; (c) a VH CDR3 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 15, 21 , 26, 32, and 39, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 15, 21 , 26, 32, and 39; (d) a VL CDR1 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 16, 22, 27, 33, and 34, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 16, 22, 27, 33, and 34; (e) a VL CDR2 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 17, 23, 28, and 35, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 17, 23, 28, and 35; and (f) a VL CDR3 region comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 18, 24, 29, and 36, or an amino acid sequence having one, two, three, four or five amino acid substitutions, deletions or additions as compared to SEQ ID NOs: 18, 24, 29, and 36.
Engineered antibodies of the invention include those in which modifications have been made to framework residues within VH and/or VL, e.g., to improve the properties of the antibody. Typically such framework modifications are made to decrease the immunogenicity of the antibody. For example, one approach is to "backmutate" one or more framework residues to the corresponding germline sequence (also referred to as "germlining"). More specifically, an antibody that has undergone somatic mutation may contain framework residues that differ from the germline sequence from which the antibody is derived. Such residues can be identified by comparing the antibody framework sequences to the germline sequences from which the antibody is derived.
Another type of framework modification involves mutating one or more residues within the framework region, or even within one or more CDR regions, to remove T cell epitopes to thereby reduce the potential immunogenicity of the antibody. This approach is also referred to as "deimmunization" and is described in further detail in U.S. Patent
Publication No. 2003/0153043. In addition to, or alternative to, modifications made within the framework or CDR regions, antibodies of the invention may be engineered to include modifications within the Fc region, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen- dependent cellular cytotoxicity. Furthermore, an antibody of the invention may be chemically modified (e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation.
Another modification of the antibodies herein that is contemplated by the invention is pegylation. An antibody can be pegylated to, for example, increase the biological (e.g., serum) half life of the antibody. To pegylate an antibody, the antibody, or fragment thereof, typically is reacted with polymers including polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment. Alternatively, the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer). As used herein, the term "polyethylene glycol" is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (C1 -C10) alkoxy- or aryloxy-polyethylene glycol or polyethylene glycol-maleimide. In certain embodiments, the antibody to be pegylated is an aglycosylated antibody. Methods for pegylating proteins are known in the art and can be applied to the antibodies of the invention.
Methods of Engineering Antibodies
As discussed above, the selective anti-tofacitinib antibodies having VH and VL sequences disclosed herein can be used to create new anti-tofacitinib antibodies by modifying the VH and/or VL sequences, or the constant region(s) attached thereto. Thus, in another aspect of the invention, the structural features of an anti-tofacitinib antibody of the invention are used to create structurally related anti-tofacitinib antibodies that retain at least one functional property of the antibodies of the invention, such as binding to tofacitinib but not substantially binding to metabolite 1 and/or metabolite 2. For example, one or more CDR regions of 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5, 12H4.G2, 16F10.E6, and/or 12D4.G6, or mutations thereof, can be combined recombinantly with known framework regions and/or other CDRs to create additional, recombinantly-engineered, anti-tofacitinib antibodies, as discussed above. Other types of modifications include those described in the previous section. The starting material for the engineering method is one or more of the VH and/or VL sequences provided herein, or one or more CDR regions thereof. To create the engineered antibody, it is not necessary to actually prepare {i.e., express as a protein) an antibody having one or more of the VH and/or VL sequences provided herein, or one or more CDR regions thereof. Rather, the information contained in the sequence(s) is used as the starting material to create a "second generation" sequence(s) derived from the original sequence(s) and then the "second generation" sequence(s) is prepared and expressed as a protein.
Standard molecular biology techniques can be used to prepare and express the altered antibody sequence.
Optionally, the antibody encoded by the altered antibody sequence(s) is one that retains one, some, or all of the functional properties of the anti-tofacitinib antibodies described herein, which functional properties include, but are not limited to:
(i) does not substantially bind to tofacitinib metabolite 1 of Formula II; and/or
(ii) does not substantially bind to tofacitinib metabolite 2 of Formula III. The functional properties of the altered antibodies can be assessed using standard assays available in the art and/or described herein, as well as those known in the art or those discovered in the future.
In certain embodiments of the methods of engineering antibodies of the invention, mutations can be introduced randomly or selectively along all or part of an anti-tofacitinib antibody coding sequence and the resulting modified anti-tofacitinib antibodies can be screened for binding activity and/or other functional properties as described herein. Mutational methods are well-known in the art.
Production of Monoclonal Antibodies of the Invention
Monoclonal antibodies (mAbs) of the present invention can be produced by a variety of techniques, including conventional monoclonal antibody methodology. Mouse, rat, rabbit, camelid and human monoclonal antibodies of the invention can also be prepared using phage display methods for screening libraries of immunoglobulin genes from those, or many other, species. Such phage display methods for isolating antibodies are established in the art and encompass phage display libraries prepared from immunized animals or humans or naive phage display libraries where the original starting material is not derived from immunized animals. Phage display is well known in the art and is described, for example, in U.S. Patent No. 5,223,409, WO 91/17271 , WO92/20791 ; and WO 92/15679.
The monoclonal antibodies of the invention can also be prepared by culturing a host cell comprising a nucleic acid encoding the monoclonal antibody under suitable conditions and recovering said antibody, or antigen binding portion thereof. Such host cell culturing methods are well-known in the art.
Methods of Use
This disclosure further provides a method for assessing the presence or concentration of tofacitinib in a sample, the method comprising providing a sample suspected of containing tofacitinib; contacting the sample or sample extract with an antibody specific for tofacitinib under conditions suitable for binding of the antibody to tofacitinib to form an assay mixture; and detecting binding of the antibody to tofacitinib.
In another embodiment, this disclosure provides a method of determining the concentration of tofacitinib in a sample. The method comprises providing a labeled competitor comprising tofacitinib coupled to a detectable label; providing a selective anti- tofacitinib antibody that binds to tofacitinib but not to metabolites 1 and/or 2; combining the sample, the selective anti-tofacitinib antibody, and the labeled competitor, wherein the tofacitinib in the sample competes with the labeled competitor for binding to the selective anti-tofacitinib antibody; and determining the concentration of tofacitinib in the sample by measuring the amount of the labeled competitor not bound to the antibody by detection of the label. In an exemplary embodiment, the amount of labeled competitor (tofacitinib) is detected in a sample that does not contain tofacitinib and the amount of label detected in the sample is compared to the amount of label present where tofacitinib may be present. The difference between the amount of label detected in the absence of tofacitinib and the amount of label in the sample that is suspected of containing tofacitinib is measured. In another embodiment, this disclosure provides competitive immunoassay kits for determining the presence or absence of tofacitinib in a sample are provided. Illustrative competitive immunoassay kits, such as an enzyme linked immunoassay (ELISA), comprise an antibody capable of specifically binding tofacitinib, and a tofacitinib compound conjugated to a detectable label, wherein the conjugated tofacitinib compound is configured to compete with the tofacitinib in the sample to bind with the antibody, and wherein the label provides a signal indicative of a concentration of tofacitinib in the sample when the tofacitinib in the sample is present in therapeutic drug monitoring concentrations.
In one embodiment, this disclosure provides a competitive immunoassay for determining the presence of tofacitinib in a sample at a concentration of drug within the therapeutic range of tofacitinib. However, it is understood that it may be useful to provide information across a broader range, and the immunoassay range is generally broader than the therapeutic range. Accordingly, immunoassays that monitor therapeutic drug concentrations may provide sensitivity across a broader range of tofacitinib concentrations. In one aspect, the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 0 to about 1500 ng/ml. In another aspect, the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 5 to about 1215 ng/ml. In another aspect, the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 5 to about 500 ng/ml. In another aspect, the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 5 to about 405 ng/ml. In another aspect, the competitive immunoassays are suitable for monitoring the presence of tofacitinib in a sample at a concentration in the range of about 15 to about 1215 ng/ml.
In one competitive immunoassay, the labeled competitor may be derived from tofacitinib using a linkage that is the same or similar to that of the immunogenic tofacitinib conjugate. In some competitive immunoassays, it is desirable to have a labeled competitor that binds to the anti-tofacitinib antibody with less specificity than tofacitinib binds to the anti- tofacitinib antibody, allowing for the labeled competitor to be displaced more readily in the presence of tofacitinib.
The antibodies, immunogenic tofacitinib conjugates, labeled competitors and/or other conjugates described herein are also suitable for use with any of a number of other homogeneous and heterogeneous immunoassays with a range of detection systems. The examples presented herein are not intended to be limiting.
Thus, the present invention provides tofacitinib conjugates that are useful for the preparation of immunogens and conjugates for use in immunoassays for the detection of tofacitinib. By coupling a tofacitinib analog according to the present invention to an immunogenic carrier material, polyclonal or monoclonal antibodies can be produced and isolated, which are useful reagents for immunoassays for the detection of tofacitinib. Coupling can be accomplished by any chemical reaction that will bind the label or carrier.
Illustrative tofacitinib immunoassays employ anti-tofacitinib antibodies that can be either polyclonal or monoclonal. In illustrative competitive immunoassays, the antibody preparation used is induced by an immunogen described herein is formulated in an aqueous solution such as buffer, and the like or provided in an adjuvant or similar composition. The induced antibodies can be tested to determine specificity for tofacitinib.
The antibodies and labeled competitors described herein are also suitable for use with any of a number of other homogeneous and heterogeneous immunoassays with a range of detection systems.
Kits
The invention provides a kit for determining the concentration of tofacitinib in a sample, the kit comprising at least one selective anti-tofacitinib antibody. In one aspect, the kit further comprises a labeled competitor comprising tofacitinib coupled to a detectable label. In another embodiment, the labeled competitor competes with the tofacitinib in a sample for binding to the anti-tofacitinib antibody. In another aspect, the selective anti- tofacitinib antibody is produced using an immunogenic tofacitinib conjugate comprising tofacitinib coupled to an immunogenic carrier. The kit may also include an applicator and/or instructional material for the use of the kit.
In another embodiment, the invention includes a kit for determining the concentration of tofacitinib in a sample wherein the kit comprises at least one selective tofacitinib antibody that binds tofacitinib but does not substantially bind at least one metabolite selected from the group consisting of tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III. In one aspect, the kit comprises at least one selective tofacitinib antibody that binds tofacitinib but does not substantially bind tofacitinib metabolite 1 of Formula II and does not substantially bind tofacitinib metabolite 2 of Formula III. In one aspect, the kit further comprises an applicator. In another aspect, the kit comprises an instructional material for the use of the kit.
In one aspect, the kit can detect a concentration of tofacitinib ranging from 5 ng/ml to 1215 ng/ml. In another aspect, the kit can detect a concentration of tofacitinib ranging from 5 ng/ml to 405 ng/ml. In yet another aspect, the kit can detect a concentration of tofacitinib ranging from 15 ng/ml to 1215 ng/ml.
The present invention is further illustrated by the following examples which should not be construed as further limiting. The contents of all figures and all references, patents and published patent applications cited throughout this application are expressly incorporated herein by reference.
EXAMPLES
EXAMPLE 1 :
Conjugation of tofacitinib with succinic acid
40 mg succinic anhydride and 0.2 ml TEA (50 mM triethanolamine, 50 mM KCI, 20 mM MgCI2, pH 7.5) were added to a solution of 47 mg of tofacitinib in 0.8 ml acetonitrile. The reaction mixture was shaken at a temperature of 60°C for one hour. At one hour, an additional 50 mg succinic anhydride was added to the reaction mixture, followed by addition one incubation at 60°C with shaking. Purified material was obtained following HPLC purification in a mobile phase gradient of 0-30% acetonitrile (ACNN) affording 25.2 mg of the tofacitinib hemisuccinate intermediate of Formula IV.
Figure imgf000036_0001
Formula IV
EXAMPLE 2:
Synthesis of an immunogenic tofacitinib conjugate comprising tofacitinib coupled to BSA
1.5 mg of the compound of Formula IV was dissolved in 2 ml 1-Ethyl-3-[3- dimethylamino-propyl]carbodiimide hydrochloride (EDC) conjugation buffer (0.1 M MES, 0.9 M NaCI, 0.02% NaN3, pH 4.7). 8 mg BSA (Imject BSA obtained from Pierce #77601 ) was dissolved in 0.8 ml water and then mixed with the formula IV solution. Next, 10 mg EDC was dissolved in 0.1 ml H20 and then immediately added to BSA, Formula IV mixture. The mixture was gently shaken for 2 hours at room temperature. The mixture was spun down and the supernatant containing the tofacitinib-BSA conjugate (Formula V) was purified by desalting according to standard desalting methods known to those of skill in the art. Conjugation of tofacitinib with BSA was confirmed by Matrix-Assisted Laser Desorption Ionization (MALDI).
Figure imgf000037_0001
N-BSA Formula V
EXAMPLE 3:
Synthesis of an immunogenic tofacitinib conjugate comprising tofacitinib coupled to BSA
1.5 mg of the compound of Formula IV was dissolved in 1.5 ml EDC conjugation buffer. 10 mg EDC was dissolved in 0.2 ml water and immediately added into Formula IV solution. The Formula IV/EDC mixture was added into 0.6 ml Mariculture Keyhole Limpet Hemocyanin (mcKLH, obtained from Pierce #77601 ) solution (10 mg/ml in water). The reaction mixture was gently shaken at room temperature for two hours. The mixture was spun down and the supernatant containing the tofacitinib-KLH conjugate (Formula VI) was purified by desalting according to standard desalting methods known to those of skill in the art.
Figure imgf000037_0002
N-KLH
EXAMPLE 4:
Synthesis of a tofacitinib labeled competitor
One illustrative example of a tofacitinib labeled competitor is a biotinylated tofacitinib derivative of Formula VII. The biotinylated tofacitinib derivative of Formula VII was produced as follows.
18 mg of the compound of Formula IV and 20 mg EDC were dissolved in 1 ml EDC conjugation buffer. 0.1 ml acetonitrile was added to help dissolve the compound. The pH of the solution was adjusted to 5.5 with addition of 10 μΙ 5 M NaOH. Then, 23 mg amine-PEG3- biotin was added. The mixture was shaken at room temperature overnight. Following the overnight incubation, biotinylated tofacitinib (Formula VII) was purified by HPLC in a mobile phase gradient of 0-30% ACN affordin 14 mg of purified compound.
Figure imgf000038_0001
Formula VII
EXAMPLE 5:
Production of Anti-tofacitinib Antibodies.
Polyclonal and monoclonal antibodies were produced by using conventional techniques, essentially as described by Kohler and Milstein, Nature 256:495-497(1975). Three female Balb/C mice were immunized with 100 μg of the immunogenic tofacitinib-KLH conjugate of example 3 in complete Freund's adjuvant, administered by intraperitoneal (ip) injection. Three subsequent injections were administered ip comprising 50 μg mouse of the immunogenic tofacitinib-KLH conjugate in incomplete Freund's adjuvant on days 10, 20, and 30 after initial injection. Serum was collected on day 37 and the presence of antibodies reactive to the antigen was determined by solid phase ELISA, as described below, against immunogenic tofacitinib-BSA conjugate of example 2.
Solid Phase ELISA: The immunogenic tofacitinib-BSA conjugate of example 2 was immobilized to wells of a microtiter plate. Specifically, microtiter plates were coated with 2- 10 μg ml tofacitinib-BSA conjugate in a coating buffer (0.2 M carbonate buffer (BuPH carbonate-bicarbonate buffer pack, Pierce item #28382) for 1-2 hours at room temperature or overnight at 4°C, then saturated with a blocking buffer (PBS containing 1 % (w/v) BSA) for 1 hour at room temperature or overnight at 4°C, and washed 3x with washing buffer (PBS containing 0.5% (v/v) Tween 20). The antibody samples (i.e., the mouse serum or the hybridoma supernatants) to be screened were diluted in a diluent (0.1 % solution of BSA in PBS). The diluted antibody samples were added to microtiter plate and the plate was incubated for 1-2 hours at room temperature. After having washed away any unbound substances with washing buffer, the level of bound anti-tofacitinib antibody was determined using rabbit or goat anti-mouse peroxidase-conjugated secondary antibody (IgG specific) at recommended or experimentally derived dilution (usually 1/1000-1/10,000) in the diluent. After incubation for 30 minutes at room temperature and 3x wash with washing buffer, the chromogenic substrate, OPD (o-phenylenediamine), was added, then color was developed for 20 minutes, and stopped by adding 50 μΙ 2 N sulfuric acid. Absorbance at 490 nm was measured. Figure 1 is a graph depicting binding of serum antibodies to the tofacitinib-BSA conjugate of example 2 as determined by solid phase ELISA. As demonstrated in Figure 1 , antibodies reactive to the antigen were present in each of mouse 1 , 2 and 3. As mouse 3 had the highest titer (defined as the dilution of serum at 50% maximum signal), this mouse was selected for hybridoma generation.
Hybridoma generation: The mouse demonstrating the highest serum antibody titer (defined as the dilution of serum at 50% maximum signal), mouse 3, received booster injection comprising 25 μg of antigen. Three days later, the mouse was sacrificed and its spleen cells were isolated and fused with NS1 myeloma cells following a standard fusion protocol. These mixtures of clones, called parental clones, were screened at 10 days after the fusion by solid-phase ELISA (described above) against immunogenic tofacitinib-BSA to identify parental clones that secreted antibodies capable of binding tofacitinib. Positive parental clones were selected for further analysis.
These positive parental clones originating from the fusion were expanded from the 96 well plates to 24 well plates and were rescreened 3 days later to ensure that the clones were still producing antibody. From the rescreen, selected wells with positive hybridomas, (i.e., hybridomas that secreted antibodies capable of binding the immunogenic tofacitinib-BSA conjugate and that did not bind to metabolite-1-BSA, metabolite-2-BSA) were frozen for future use and sub-cloned to isolate positive cell lines.
The selected positive parent clones that secreted antibodies capable of binding immunogenic tofacitinib-BSA conjugate were subcloned by limiting dilution to obtain monoclonal hybridoma cell lines. Approximately 10 days after subcloning, small volumes of media were removed from the wells and screened by solid-phase ELISA to identify antibody- producing clones. Selected positive clones were expanded and rescreened to ensure that the clones were still producing antibody and confirm specificity. Up to two positive clones per parental line, including clones 6D9.A5 and 12D4.G6, were expanded for freezing 2 vials each. 1 ml supernatant was also collected for testing.
In addition to the direct solid-phase ELISA described above in Example 5, alternative immunoassay formats were developed, including competitive a competitive immunoassay format for the detection of tofacitinib in experimental samples. Exemplary competitive immunoassay protocols are provided below.
Given that tofacitinib is a powerful immunosuppressant, the production of antibodies to tofacitinib was surprising. Even more surprising, was the exquisite selectivity of these antibodies in that, as shown below, they distinguished between tofacitinib and two metabolites thereof, selectively binding tofacitinib but not substantially binding either one or both metabolites. Competitive Anti-tofacitinib lmmunoassay-1
The direct solid-phase ELISA described above in Example 5 was converted to a competitive ELISA wherein a defined amount of monoclonal antibody replaced immune serum samples or hybridoma supernatants, a competitor (i.e., tofacitinib, tofacitinib metabolites) was then added to the monoclonal antibody solution, and binding of the monoclonal antibody to the tofacitinib-BSA conjugate in the presence and absence of the competitor was measured.
Competitive Anti-tofacitinib Immunoassay -2
Microtiter plates were coated with 100 μΙ/well 1 μg ml purified anti-tofacitinib antibody in a coating buffer for 3 hours at room temperature, then replaced with 300 μΙ/well blocking buffer (PBS containing 1 % non-fat dry milk powder) for 30 minutes at room temperature, and washed 3x with washing buffer (PBS containing 0.5%) Tween 20). 100 μΙ/well of mixture biotinylated tofacitinib (0.03 μg ml), an unlabeled competitor, such as tofacitinib (1 .0-0.001 μg ml), Metabolite 1 or Metabolite 2, and streptavidin-HRP (1 :16,000 dilution, may vary with lot) was added and incubated for 2 hours at room temperature. After 3x washing, 100 μΙ OPD substrate was added, then color was developed for 20 minutes, and the reaction was stopped by adding 50 μΙ 2 N sulfuric acid. Absorbance at 450 nm was measured.
Competitive Anti-tofacitinib Immunoassay -3
200 μΙ/well of mixture containing anti-tofacitinib MAb (1/100 to 1/1638400 dilution), biotinylated tofacitinib (1 ng/ml), and a non-labeled competitor, such as tofacitinib (1000 to 0.001 ng/ml), Metabolite 1 (400 - 0.097 ng/ml) or Metabolite 2, was added into goat anti- mouse IgG pre-coated plates, and incubated for 1-2 hour at room temperature. After washing the plates, 200 μΙ of streptavidin-HRP (50 ng/ml) was added and the plates were then incubated for 0.5-1 hour at room temperature. After washing the plates again, 200 μΙ of TMB (3,3',5,5'-Tetramethylbenzidine) substrate was added, then color developed for 30 minutes, and the reaction was stopped by adding 50 μΙ 2 N sulfuric acid. Absorbance at 450 nm was measured.
EXAMPLE 6:
Binding of Anti-tofacitinib Monoclonal Antibodies to Biotinylated tofacitinib Selected antibodies were further tested for binding to tofacitinib by standard ELISA. Briefly, a total of 34 clones were screened for binding to biotinylated tofacitinib. The format of the ELISA was as follows. Biotinylated tofacitinib (1 ng/mL) was incubated with antibody dilutions ranging from 1 :100 to 1 :1 ,638,400 on a pre-coated goat anti-mouse IgG plate for one hour. After having washed away any unbound substances, the binding of biotinylated tofacitinib to antibody was measured after the addition of streptavidin-HRP and the related chromogenic substrate TMB according to known methods. Figure 2 is a graph depicting binding of selected anti-tofacitinib antibodies to biotinylated tofacitinib as determined by ELISA. Eleven antibodies were found to bind to biotinylated tofacitinib in a concentration dependent manner. Data for ten of these antibodies are shown in Figure 2 and the data for antibody 8B5.F2 is not shown.
These eleven clones were selected for further analysis. Specifically, these eleven clones were evaluated for cross reactivity with the tofacitinib metabolite 1.
EXAMPLE 7:
Binding of Anti-tofacitinib Monoclonal Antibodies to Metabolite 1 Antibodies were be further characterized as binding to tofacitinib or its metabolites by measuring cross reactivity using competitive immunoassay-3 mentioned above wherein tofacitinib metabolites as competitors.
For this example, eleven anti-tofacitinib monoclonal antibodies were screened for cross reactivity with metabolite 1 using competitive immunoassay. 200 μΙ/well of mixture containing anti-tofacitinib MAb(1/100 to 1/1638400 dilution), biotinylated tofacitinib (1 ng/ml), and Metabolite 1 (400 - 0.097 ng/ml), was added into goat anti-mouse IgG pre-coated plates, and incubated for 1 -2 hour at room temperature. After washing the plates, 200 μΙ of streptavidin-HRP (50 ng/ml) was added and then incubated for 0.5-1 hour at room temperature. After washing the plates again, 200 μΙ of TMB substrate was added, then developed for 30 minutes, and stopped by adding 50 μΙ 2 N sulfuric acid. Absorbance at 450 nm was measured.
Figure 3 is a graph depicting cross reactivity of selected anti-tofacitinib antibodies to metabolite 1 as determined using the competitive immunoassay. As demonstrated in Figure 3, four antibodies demonstrated cross reactivity to metabolite 1 in a concentration dependent manner (e.g., clones 7C10.C1 1 , 1 B2.B9, 1 1 G10.F12, and 8B5.F2). Seven clones were selective in that they bound tofacitinib but did not detectably bind metabolite 1 , e.g., 5A3.E5, 10A6.C5, 10F10.H5, 6D9.A5 (or 6D9.A8), 12H4.G2, 16F10.E6, and 12D4.G6. These plots for the seven clones are shown as the lines going approximately straight across the top of the graph depicted in Figure 3. More specifically, the data used to prepare the graph shown in Figure 3 are as follows:
TABLE 1
Figure imgf000041_0001
Figure imgf000042_0001
The seven clones that did not show cross reactivity with Metabolite 1 were further tested for inhibition with the parent drug.
The amino acid sequences of the heavy and light chain variable domains of the seven monoclonal antibody clones that did not show cross reactivity with Metabolite-1 are provided in Table 2 below.
TABLE 2
Figure imgf000043_0001
Antibody Heavy Chain V domain (CDRa (bold)) Light Chain V domain (CDR (bold))
CDR1 GYSITSGYS (SEQ ID NO:19) Q VGTN (SEQ ID NO:34)
CDR2 IHYSGNT (SEQ ID NO:20) SAS (SEQ ID NO:35)
CDR3 VRGPYGSSFY (SEQ ID NO:21 ) QQYYNYPLT (SEQ ID NO:36)
12D4.G6 QVQLKESGPGLVAPSQSLSITCTVSG NIVMTQSPKSMSMSVGERVTLTCKAS
FSLSIYSVHWVROPPGKGLEWLGMIW E WTYVSWYOOKPEOSPKLLIYGAS GGGNTDYNSVLKSRLSISKDNSKSOV NRYTGVPDRFTGSGSATDFTL ISSV FLKV SLOTDDTAMYYCARIYYGIFW OAEDLADYHCGQGYSYPYTFGGGTKL GQGTLVTVSA (SEQ ID NO: 12) EIKR (SEQ ID NO:4)
CDR1 GFSLSIYS (SEQ ID NO:37) E WTY (SEQ ID NO:22)
CDR2 IWGGGNT (SEQ ID NO:38) GAS (SEQ ID NO:23)
CDR3 ARIYYGIF (SEQ ID NO:39) GQGYSYPYT (SEQ ID NO:24) aThe Complementarity Determining Regions (CDRs) are highlighted in BOLD and underline as determined by the IMGT numbering system (Lefranc, M.-P. et al., 1999, Nucleic Acids Research 27:209-212).
b Antibody clones 6D9.A8 and 16F10.E2 failed to survive the archival process. For this reason, clones 6D9.A5 and 16F10.E6, subcloned from the same parental lines, were sequenced.
EXAMPLE 8:
Antigen Competition
For this example, a competitive immunoassay format was used to determine whether tofacitinib inhibited the ability of the selected antibody clones that did not cross react with Metabolite- 1 to bind biotinylated tofacitinib. The selected antibody clones that did not cross react with Metabolite-1 were assayed at their EC50 dilution, determined from the initial antibody titration (see Figure 2). 200 μΙ/well of mixture containing anti-tofacitinib MAb (1/100 to 1/1638400 dilution), biotinylated tofacitinib (1 ng/ml), and unlabeled tofacitinib (1000 to 0.001 ng/ml) was added into goat anti-mouse IgG pre-coated plates, and incubated for 1 -2 hour at room temperature. After washing the plates, 200 μΙ of streptavidin-HRP (50 ng/ml) was added and then incubated for 0.5-1 hour at room temperature. After washing the plates again, 200 μΙ of TMB substrate was added, then developed for 30 minutes, and stopped by adding 50 μΙ 2N sulfuric acid. Absorbance at 450 nm was measured.
As demonstrated in Table 3 below, competition was seen with clones 6D9.A8 and 12D4.G6. Specifically, inhibition of binding to biotinylated tofacitinib by unlabeled tofacitinib was observed with clones 6D9.A8 and 12D4.G6, with an ED50 of 202 ng/mL and 104 ng/mL, respectively. Table 3 below shows the ED20, ED50 and ED80 for this experiment. TABLE 3.
Antigen Competition Assay Results
Figure imgf000045_0001
EXAMPLE 9:
Binding of Anti-tofacitinib Monoclonal Antibodies to Metabolite 1 and Metabolite 2: Two clones, 6D9.A8 and 12D4.G6, were tested for cross reactivity to metabolites 1 and 2. In this experiment, the metabolites were prepared at concentrations of 200,000 to 200 ng/ml and the antibody clones were prepared at their EC50 dilutions shown in Table 1 . The metabolites were incubated with biotinylated tofacitinib (1 ng/mL) and antibody on a pre- coated goat anti-mouse IgG plate for one hour. After having washed away any unbound substances, the binding of biotinylated tofacitinib to antibody was measured after the addition of streptavidin-HRP and the related chromogenic substrate TMB according to known methods.
Table 4 below summarizes the cross reactivity of clones 6D9.A8 and 12D4.G6 to Metabolite-1 and Metabolite-2.
TABLE 4
Clone cross reactivity to Metabolite 1 and Metabolite 2
Figure imgf000045_0002
As shown in Table 4, clone 6D9.A8 showed 3.68% and 0.38% cross reactivity to Metabolites 1 and 2, respectively; and clone 12D4.G6 showed 4.17% and 0.15% cross reactivity to Metabolites 1 and 2, respectively. These data demonstrate that these antibodies do not substantially bind to metabolites 1 and 2.
EXAMPLE 10:
Standard Curve with Tofacitinib
For this example, the relative affinity of a selected antibody to tofacitinib was determined using solutions containing known concentration of labeled tofacitinib (e.g., 0.001 to 1000 ng/ml). Figure 4 is a graph depicting optimal dilution ranges of select monoclonal antibodies of the invention, e.g., 6D9.A8 (open circles) and 12D4.G6 (open squares) as determined by ELISA. Table 5 summarizes the assay conditions and results of the ELISA. Table 6 shows the data values used to generate the graph shown in Figure 5.
TABLE 5
Figure imgf000046_0001
Figure imgf000046_0002
Curve Fit Option - Fixed Weight Value. Subclones 6D9.A8 and 12D4.G6 demonstrated similar dose responses to the parent drug with 6D9.A8 being slightly more potent.
As demonstrated in Figure 4 and Table 5, clone 12D4.G6 showed standard range of 5 to 405 ng/ml (Figure 4), demonstrating that this monoclonal antibody is capable of detecting tofacitinib at levels as low as 5 ng/ml. Figure 4 also showed standard range of 6D9.A8 was 15 - 1215 ng/ml. Thus, combined use of these two antibodies provides a dose range for detection of tofacitinib in a sample of from about 5 ng/ml to about 1215 ng/ml.
EXAMPLE 1 1 :
Spike and recovery in heparinized human plasma
For this example, tofacitinib was spiked into commercially available heparin human plasma to determine the effect of dilution on linearity and recovery of tofacitinib in human plasma sample. Tofacitinib was spiked into Heparin-plasma (human) at a concentration of 4864 ng/ml and serially diluted 2-fold to test assay tolerance to plasma. Percent dilution linearity and percent recovery was calculated via an ELISA assay as described in Example 5. TABLE 7
The effect of dilution on linearity and recovery of tofacitinib in human plasma sample.
Figure imgf000047_0001
aLOD = level o detection
As Table 7 demonstrates, plasma samples obtained from a commercial source that were diluted at least 1 :8 in assay buffer had acceptable dilution linearity and recovery. Samples diluted 2- and 4-fold returned lower than expected absorbance values, indicating sample matrix interference at these lower dilutions. These data demonstrate that the antibodies of the invention can accurately detect presence and concentration of tofacitinib even in a complex biological sample such as, but not limited to, plasma.
EXAMPLE 12:
Clone 6D9.A5 characterization
For this example, tofacitinib dose response curves (0.001 to 1000 ng/ml) were run in the assay using antibody clones 6D9.A5, 6D9.A8, or 12D4.G6. The results, are shown in the Table 8 and Figure 5. Further, the data used to generate the graph shown in Figure 5 are set forth below in Table 9. The data disclosed herein indicate that 6D9.A5 has same standard range (15-1215 ng/ml) as 6D9.A8 and is capable of detecting tofacitinib in a sample at concentrations as low as about 15 ng/ml and as high as about 1215 ng/ml.
TABLE 8
Figure imgf000047_0002
TABLE 9
Figure imgf000048_0001
Curve Fit Option - Fixed Weight Value.
Thus, the replacement subclone antibody 6D9.A5 (replacing 6D9.A8) demonstrated a similar dose response activity to the parent drug as compared to subclone antibody 6D9.A8. In addition, the data disclosed herein demonstrate that 6D9.A5 and 6D9.A8 antibodies are similarly more potent binders of the parent drug compared with antibody 12D4.G6.
6D9.A5 antibody was further characterized by measuring cross reactivity to the metabolites using competitive immunoassay-3, wherein tofacitinib metabolites were used as competitors or using direct ELISA assay mentioned in Example 5, wherein the microtiter plates were coated with tofacitinib-BSA conjugate. The data shown in Table 10 demonstrate that clone 6D9.A5 had similar cross reactivity to the metabolites as observed with clone 6D9.A8 and clone 12D4.G6. That is, like 12D4.G6, 6D9.A5 did not substantially bind metabolite 1 or 2.
TABLE 10
Figure imgf000048_0002
Although the disclosed teachings have been described with reference to various applications, methods, kits, and compositions, it will be appreciated that various changes and modifications can be made without departing from the teachings herein and the claimed invention below. The foregoing examples are provided to better illustrate the disclosed teachings and are not intended to limit the scope of the teachings presented herein. While the present teachings have been described in terms of these exemplary embodiments, the skilled artisan will readily understand that numerous variations and modifications of these exemplary embodiments are possible without undue experimentation. All such variations and modifications are within the scope of the current teachings.
All references cited herein, including patents, patent applications, papers, text books, and the like, and the references cited therein, to the extent that they are not already, are hereby incorporated by reference in their entirety. In the event that one or more of the incorporated literature and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls.
The foregoing description and Examples detail certain specific embodiments of the invention and describes the best mode contemplated by the inventors. It will be appreciated, however, that no matter how detailed the foregoing may appear in text, the invention may be practiced in many ways and the invention should be construed in accordance with the appended claims and any equivalents thereof.

Claims

WHAT IS CLAIMED IS:
1 . An immunogenic tofacitinib conjugate comprising tofacitinib coupled to an
immunogenic carrier, wherein the immunogenic carrier is a protein selected from the group consisting of keyhole limpet hemocyanin and bovine serum albumin, and further wherein
(a) the immunogenic tofacitinib conjugate has the structure
Figure imgf000050_0001
N-BSA
Formula V
wherein BSA is bovine serum albumin; or
(b) the immunogenic tofacitinib conjugate has the structure
Figure imgf000050_0002
N-KLH
Formula VI
wherein KLH is keyhole limpet hemocyanin.
2. A method for assessing the concentration of tofacitinib in a sample, the method comprising:
(a) providing a sample suspected of containing tofacitinib;
(b) contacting the sample or sample extract with an antibody specific for tofacitinib under conditions suitable for binding of the antibody to tofacitinib to form an assay mixture; and
(c) detecting binding of the antibody to tofacitinib.
3. An isolated antibody having greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
Figure imgf000051_0001
Formula II Formula III.
4. The isolated antibody of claim 3, wherein the antibody has a binding affinity for tofacitinib that is 25, 30, 40, or 50 times the binding affinity for metabolite 1 or metabolite 2.
5. The isolated antibody of claim 4, where the antibody is obtained using an
immunogenic tofacitinib conjugate compound comprising tofacitinib coupled to an immunogenic carrier.
6. A method of determining the amount of tofacitinib in a sample, the method
comprising:
(a) providing known amount of a labeled competitor comprising tofacitinib coupled to a detectable label;
(b) providing a selective anti-tofacitinib antibody;
(c) combining the sample, the selective anti-tofacitinib antibody and the labeled competitor, wherein the tofacitinib in the sample competes with the labeled competitor for binding to the selective anti-tofacitinib antibody; and
(d) determining the amount of tofacitinib in the sample by measuring the amount of labeled competitor not bound to antibody by detection of the label.
7. The method of claim 6, wherein the selective anti-tofacitinib antibody has a greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II
Figure imgf000052_0001
Formula II
and/or f metabolite 2 of Formula III
Figure imgf000052_0002
Formula III.
8. The method of claim 7, wherein the antibody has greater binding affinity for
tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of Formula III
Figure imgf000052_0003
Formula II Formula III,
9. A kit for determining the amount of tofacitinib in a sample, the kit comprising: a labeled competitor comprising tofacitinib coupled to a detectable label; and at least one selective anti-tofacitinib antibody;
wherein the labeled competitor competes with the tofacitinib in a sample for binding to the anti-tofacitinib antibody.
10. The kit of claim 9, wherein the selective anti-tofacitinib antibody has a greater binding affinity for tofacitinib than for at least one tofacitinib metabolite selected from the group consisting of tofacitinib metabolite 1 of Formula II
Figure imgf000053_0001
Formula II
and t III
Figure imgf000053_0002
Formula III.
1 1. The kit of claim 10, wherein the antibody has greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of
Formula III
Figure imgf000053_0003
Formula II Formula III.
12. A competitive immunoassay kit for determining the concentration of tofacitinib in a sample, the competitive immunoassay comprising
(a) at least one selective anti-tofacitinib antibody;
(b) a tofacitinib compound conjugated to a detectable label, wherein the conjugated tofacitinib compound competes with the tofacitinib in the sample to bind with the antibody; and
wherein the label provides a signal indicative of the concentration of tofacitinib in the sample when the tofacitinib in the sample is present in a therapeutic drug monitoring concentration.
13. The kit of claim 12, wherein the antibody has greater binding affinity for tofacitinib than for tofacitinib metabolite 1 of Formula II and tofacitinib metabolite 2 of
Formula III
Figure imgf000054_0001
Formula II Formula III.
14. An isolated antibody that binds to tofacitinib but does not substantially bind to at least one tofacitinib metabolite selected from the group consisting of a tofacitinib metabolite 1 of Formula II and a tofacitinib metabolite 2 of Formula III
Figure imgf000054_0002
Formula II Formula III.
15. The isolated antibody of claim 14, wherein the antibody does not substantially bind a tofacitinib metabolite 1 of Formula II and a tofacitinib metabolite 2 of Formula III
Figure imgf000055_0001
Formula II Formula III.
16. The antibody of claim 3, wherein the antibody can detect tofacitinib in a sample but not substantially detect a tofacitinib metabolite in the sample, and wherein the amount of tofacitinib ranges from about 5 ng/mL to 1215 ng/mL, from about 5 ng/mL to 405 ng/mL, or about 15 ng/mL to 1215 ng/mL.
17. An isolated antibody having greater binding affinity for tofacitinib than for at least one tofacitinib metabolite selected from the group consisting of a tofacitinib metabolite 1 of Formula II and a tofacitinib metabolite 2 of Formula III, the antibody comprising a heavy chain variable domain and a light chain variable domain, wherein the heavy chain variable domain comprises three complementarity determining regions (HCDR1 , HCDR2, and HCDR3) selected from the group consisting of:
(a) a HCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:13, SEQ ID NO:19, SEQ ID NO:30, and SEQ ID NO:37;
(b) a HCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:14, SEQ ID NO:20, SEQ ID NO:25, SEQ ID NO:31 , and SEQ ID NO:38;
(c) a HCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:15, SEQ ID NO:21 , SEQ ID NO:26, SEQ ID NO:32, and SEQ ID NO:39;
and wherein the light chain variable domain comprises three CDRs (LCDR1 , LCDR2, and LCDR3) selected from the group consisting of:
(d) a LCDR1 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:16, SEQ ID NO:22, SEQ ID NO:27, SEQ ID NO:33, and SEQ ID NO:34; (e) a LCDR2 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:17, SEQ ID NO:23, SEQ ID NO:28, and SEQ ID NO:35; and
(f) a LCDR3 comprising an amino acid sequence selected from the group consisting of SEQ ID NO:18, SEQ ID NO:24, SEQ ID NO:29, and SEQ ID NO:34.
18. The antibody of claim 17, wherein the antibody comprises
(a) a HCDR1 comprising the amino acid sequence of SEQ ID NO:13, a HCDR2 comprising the amino acid sequence of SEQ ID NO:14, a HCDR3 comprising the amino acid sequence of SEQ ID NO:15, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:16, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18;
(b) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(c) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:25, a HCDR3 comprising the amino acid sequence of SEQ ID NO:26, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:27, a LCDR2 comprising the amino acid sequence of SEQ ID NO:28, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:29;
(d) a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(e) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:33, a LCDR2 comprising the amino acid sequence of SEQ ID NO:17, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:18;
(f) a HCDR1 comprising the amino acid sequence of SEQ ID NO:19, a HCDR2 comprising the amino acid sequence of SEQ ID NO:20, a HCDR3 comprising the amino acid sequence of SEQ ID NO:21 , and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:34, a LCDR2 comprising the amino acid sequence of SEQ ID NO:35, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:36;
(g) a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(h) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:1 , SEQ I D NO:3, SEQ ID NO:5, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:10 and SEQ ID NO:12, and further comprising a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:2, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:9, and SEQ ID NO:1 1 ;
(i) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:1 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:2;
(j) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:3 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(k) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(I) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(m) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:5 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:6; (n) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:8 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:9;
(o) a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:10 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:1 1 ;
(p) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID N03, SEQ ID NO:7 and SEQ ID NO:12 and a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4; and
(q) a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:8, SEQ ID NO:10, and SEQ ID NO:12 and a light chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:4, SEQ ID NO:9 and SEQ ID NO:1 1 .
19. The antibody of claim 15, wherein the antibody is selected from the group
consisting of:
(a) an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:30, a HCDR2 comprising the amino acid sequence of SEQ ID NO:31 , a HCDR3 comprising the amino acid sequence of SEQ ID NO:32, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24:
(b) an antibody comprising a HCDR1 comprising the amino acid sequence of SEQ ID NO:37, a HCDR2 comprising the amino acid sequence of SEQ ID NO:38, a HCDR3 comprising the amino acid sequence of SEQ ID NO:39, and further comprising a LCDR1 comprising the amino acid sequence of SEQ ID NO:22, a LCDR2 comprising the amino acid sequence of SEQ ID NO:23, and a LCDR3 comprising the amino acid sequence of SEQ ID NO:24;
(c) an antibody comprising a heavy chain variable domain comprising an amino acid sequence selected from the group consisting of SEQ ID NO:3, SEQ ID NO:7 and SEQ ID NO:12 and further comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4;
(d) an antibody comprising a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:7 and further comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4; and (e) an antibody comprising a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO:12 and further comprising a light chain variable domain comprising the amino acid sequence of SEQ ID NO:4.
20. The antibody of claim 19(a), wherein the antibody is capable of detecting tofacitinib in a sample at a concentration ranging from about 15 ng/ml to 1215 ng/ml.
21 . The antibody of claim 19(b) wherein the antibody is capable of detecting tofacitinib in a sample at a concentration ranging from about 5 ng/ml to 405 ng/ml.
22. The kit of claim 1 1 , wherein the kit comprises the antibody of claim 19(a) and the antibody of claim 19(b), wherein the kit can be used to detect tofacitinib in a sample at a concentration ranging from about 5 ng/ml to about 1215 ng/ml.
23. A nucleic acid encoding an antibody of claim 18.
24. A host cell comprising the nucleic acid of claim 23.
25. A method of producing the antibody of 18, comprising culturing the host cell of claim 24 under conditions wherein the antibody is produced.
26. The method of claim 25, further comprising isolating the antibody.
PCT/IB2013/055008 2012-06-28 2013-06-18 Anti-tofacitinib antibodies and uses thereof for drug monitoring WO2014001967A1 (en)

Priority Applications (12)

Application Number Priority Date Filing Date Title
BR112014032916A BR112014032916A2 (en) 2012-06-28 2013-05-18 anti-drug antibodies and their uses for drug monitoring
AU2013282863A AU2013282863A1 (en) 2012-06-28 2013-06-18 Anti-tofacitinib antibodies and uses thereof for drug monitoring
MX2014015088A MX2014015088A (en) 2012-06-28 2013-06-18 Anti-tofacitinib antibodies and uses thereof for drug monitoring.
KR1020147036168A KR20150014996A (en) 2012-06-28 2013-06-18 Anti-tofacitinib antibodies and uses thereof for drug monitoring
CN201380034143.8A CN104411336A (en) 2012-06-28 2013-06-18 Anti-tofacitinib antibodies and uses thereof for drug monitoring
US14/411,356 US9408922B2 (en) 2012-06-28 2013-06-18 Anti-tofacitinib antibodies and uses thereof for drug monitoring
RU2014146707A RU2014146707A (en) 2012-06-28 2013-06-18 ANTIBODIES AGAINST TOFACITINIBE AND THEIR APPLICATION FOR MEDICINAL MONITORING
JP2015519413A JP2015527987A (en) 2012-06-28 2013-06-18 Anti-tofacitinib antibody and its use for drug monitoring
EP13752676.0A EP2866840A1 (en) 2012-06-28 2013-06-18 Anti-tofacitinib antibodies and uses thereof for drug monitoring
CA2873192A CA2873192A1 (en) 2012-06-28 2013-06-18 Anti-tofacitinib antibodies and uses thereof for drug monitoring
IL236291A IL236291A0 (en) 2012-06-28 2014-12-15 Anti-tofacitinib antibodies and uses thereof for drug monitoring
HK15108023.7A HK1207309A1 (en) 2012-06-28 2015-08-19 Anti-tofacitinib antibodies and uses thereof for drug monitoring

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261665361P 2012-06-28 2012-06-28
US61/665,361 2012-06-28

Publications (1)

Publication Number Publication Date
WO2014001967A1 true WO2014001967A1 (en) 2014-01-03

Family

ID=49029140

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2013/055008 WO2014001967A1 (en) 2012-06-28 2013-06-18 Anti-tofacitinib antibodies and uses thereof for drug monitoring

Country Status (13)

Country Link
US (1) US9408922B2 (en)
EP (1) EP2866840A1 (en)
JP (1) JP2015527987A (en)
KR (1) KR20150014996A (en)
CN (1) CN104411336A (en)
AU (1) AU2013282863A1 (en)
BR (1) BR112014032916A2 (en)
CA (1) CA2873192A1 (en)
HK (1) HK1207309A1 (en)
IL (1) IL236291A0 (en)
MX (1) MX2014015088A (en)
RU (1) RU2014146707A (en)
WO (1) WO2014001967A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2018529681A (en) * 2015-09-10 2018-10-11 ベクトン・ディキンソン・アンド・カンパニーBecton, Dickinson And Company Cyclophosphamide analogues used as immunogens and assay conjugates for cyclophosphamide and ifosfamide immunoassays
US11782042B2 (en) 2017-09-21 2023-10-10 Becton, Dickinson And Company Hazardous contaminant collection kit and rapid testing
US11860173B2 (en) 2019-01-28 2024-01-02 Becton, Dickinson And Company Hazardous contaminant collection device with integrated swab and test device

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL265321B2 (en) * 2016-09-14 2024-01-01 Teneobio Inc Cd3 binding antibodies
CN107602569A (en) * 2017-10-23 2018-01-19 上海博悦生物科技有限公司 A kind of new pyrrole simultaneously [2,3 d] pyrimidine compound and its preparation method and use
TW202108628A (en) 2019-06-14 2021-03-01 美商泰尼歐生物公司 Multispecific heavy chain antibodies binding to cd22 and cd3
CN116396392B (en) * 2023-01-17 2023-10-27 珠海重链生物科技有限公司 Antibody specific to digoxigenin and related application thereof

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991017271A1 (en) 1990-05-01 1991-11-14 Affymax Technologies N.V. Recombinant library screening methods
WO1992015679A1 (en) 1991-03-01 1992-09-17 Protein Engineering Corporation Improved epitode displaying phage
WO1992020791A1 (en) 1990-07-10 1992-11-26 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US20030153043A1 (en) 1997-05-21 2003-08-14 Biovation Limited Method for the production of non-immunogenic proteins

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9307491D0 (en) * 1993-04-08 1993-06-02 Sandoz Ltd Organic compounds
KR20120041739A (en) * 2002-07-18 2012-05-02 사이토스 바이오테크놀로지 아게 Hapten-carrier conjugates and uses thereof
UY33492A (en) * 2010-07-09 2012-01-31 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5585089A (en) 1988-12-28 1996-12-17 Protein Design Labs, Inc. Humanized immunoglobulins
US5693762A (en) 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US6180370B1 (en) 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
WO1991017271A1 (en) 1990-05-01 1991-11-14 Affymax Technologies N.V. Recombinant library screening methods
WO1992020791A1 (en) 1990-07-10 1992-11-26 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
WO1992015679A1 (en) 1991-03-01 1992-09-17 Protein Engineering Corporation Improved epitode displaying phage
US20030153043A1 (en) 1997-05-21 2003-08-14 Biovation Limited Method for the production of non-immunogenic proteins

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 877 - 883
CHOTHIA; LESK, J MOL BIOL, vol. 196, no. 4, 1987, pages 901 - 917
CLEMENTI M E ET AL: "ANTIBODIES AGAINST SMALL MOLECULES", ANNALI DELL ISTITUTO SUPERIORE DI SANITA, ROME, IT, vol. 27, no. 1, 1 January 1991 (1991-01-01), pages 139 - 144, XP000673786, ISSN: 0021-2571 *
FELLOUSE ET AL., J. MOL. BIOL., vol. 373, no. 4, 2007, pages 924 - 940
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
JAYASENA, S.D., CLIN. CHEM., vol. 45, 1999, pages 1628 - 1650
JONES ET AL., NATURE, vol. 321, 1986, pages 522 - 525
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1992, NIH
KOHLER; MILSTEIN, NATURE, vol. 256, 1975, pages 495 - 497
LEFRANC, M.-P. ET AL., NUCLEIC ACIDS RES., vol. 27, 1999, pages 209 - 212
MACCALLUM ET AL., J. MOL. BIOL., vol. 262, 1996, pages 732 - 745
MAKABE ET AL., J. BIOL. CHEM., vol. 283, 2008, pages 1156 - 1166
MAKABE ET AL., JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 283, 2008, pages 1156 - 1166
MARK E FLANAGAN ET AL: "Discovery of CP-690,550: A Potent and Selective Janus Kinase (JAK) Inhibitor for the Treatment of Autoimmune Diseases and Organ Transplant Rejection", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 53, no. 24, 23 December 2010 (2010-12-23), pages 8468 - 8487, XP008153299, ISSN: 0022-2623, DOI: 10.1021/JM1004286 *
MATSUMOTO N ET AL: "Preparation of antibodies against a novel immunosuppressant, FTY720, and development of an enzyme immunoassay for FTY720", BIOORGANIC & MEDICINAL CHEMISTRY, PERGAMON, GB, vol. 14, no. 12, 15 June 2006 (2006-06-15), pages 4182 - 4192, XP027992688, ISSN: 0968-0896, [retrieved on 20060615] *
PANIAGUA RICARDO ET AL: "Quantitative analysis of the immunosuppressant CP-690,550 in whole blood by column-switching high-performance liquid chromatography and mass spectrometry detection.", THERAPEUTIC DRUG MONITORING OCT 2005, vol. 27, no. 5, October 2005 (2005-10-01), pages 608 - 616, XP008165584, ISSN: 0163-4356 *
QUEEN ET AL., PROC. NATL. ACAD. SEE. U.S.A., vol. 86, 1989, pages 10029 - 10033
RIECHMANN ET AL., NATURE, vol. 332, 1998, pages 323 - 327
TAMURA K ET AL: "A HIGHLY SENSITIVE METHOD TO ASSAY FK-506 LEVELS IN PLASMA", TRANSPLANTATION PROCEEDINGS, ELSEVIER INC, ORLANDO, FL; US, vol. 19, no. 5, 1 October 1987 (1987-10-01), pages 23 - 29, XP000973721, ISSN: 0041-1345 *
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2018529681A (en) * 2015-09-10 2018-10-11 ベクトン・ディキンソン・アンド・カンパニーBecton, Dickinson And Company Cyclophosphamide analogues used as immunogens and assay conjugates for cyclophosphamide and ifosfamide immunoassays
EP3347099A4 (en) * 2015-09-10 2019-01-30 Becton, Dickinson and Company Cyclophosphamide analogs for use as immunogens and assay conjugates for an immunoassay of cyclophosphamide and ifosfamide
US10434160B2 (en) 2015-09-10 2019-10-08 Becton, Dickinson And Company Cyclophosphamide analogs for use as immunogens and assay conjugates for an immunoassay of cyclophosphamide and ifosfamide
US11413342B2 (en) 2015-09-10 2022-08-16 Becton, Dickinson And Company Cyclophosphamide analogs for use as immunogens and assay conjugates for an immunoassay of cyclophosphamide and ifosfamide
US11782042B2 (en) 2017-09-21 2023-10-10 Becton, Dickinson And Company Hazardous contaminant collection kit and rapid testing
US11860173B2 (en) 2019-01-28 2024-01-02 Becton, Dickinson And Company Hazardous contaminant collection device with integrated swab and test device

Also Published As

Publication number Publication date
HK1207309A1 (en) 2016-01-29
AU2013282863A1 (en) 2014-11-20
US20150337054A1 (en) 2015-11-26
JP2015527987A (en) 2015-09-24
CA2873192A1 (en) 2014-01-03
MX2014015088A (en) 2015-03-05
US9408922B2 (en) 2016-08-09
RU2014146707A (en) 2016-08-20
EP2866840A1 (en) 2015-05-06
KR20150014996A (en) 2015-02-09
CN104411336A (en) 2015-03-11
IL236291A0 (en) 2015-02-26
BR112014032916A2 (en) 2017-08-01

Similar Documents

Publication Publication Date Title
JP7317272B2 (en) TIGIT Antibodies, Antigen-Binding Fragments Thereof, and Medical Uses Thereof This application is based on and claims priority from Application No. CN201710908565.3 filed on September 29, 2019. The disclosure of which is incorporated herein by reference in its entirety.
US9408922B2 (en) Anti-tofacitinib antibodies and uses thereof for drug monitoring
WO2014016737A1 (en) Novel chicken monoclonal antibodies against human phosphorylated tau and uses thereof
US11236155B2 (en) Antibodies to pyroglutamate amyloid-β and uses thereof
AU2017355596A1 (en) Antibodies to pyroglutamate amyloid-β and uses thereof
CA2964179C (en) Monoclonal anti-gpc-1 antibodies and uses thereof
JP2007528868A (en) Anti-Lewis Y anti-idiotype antibody and use thereof
WO2021112185A1 (en) Anti-gdf15 antibody
US10584175B2 (en) FN14-binding proteins and uses thereof
KR20200099421A (en) An antibody specifically binding symmetric dimethylarginine and uses thereof
JP4037933B2 (en) Anti-human calcitonin monoclonal antibody
EP2484694A1 (en) Monoclonal antibody against human hig-1 polypeptide
WO2024012434A1 (en) Antibody, antigen-binding fragment thereof, and pharmaceutical use thereof
US20220220220A1 (en) Antibodies and antigen binding peptides for factor xia inhibitors and uses thereof
US20220049018A1 (en) Anti-naloxone and anti-naltrexone monoclonal antibodies and methods of production and use thereof
JPWO2005054296A1 (en) Antibody recognizing methyllysine, method for producing the same and use thereof
EP3665203A1 (en) Method for determining anti-drug antibodies in a minipig sample
JP2020007315A (en) Monoclonal anti-gpc-1 antibodies and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13752676

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2873192

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2013282863

Country of ref document: AU

Date of ref document: 20130618

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2014/015088

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 20147036168

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2015519413

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14411356

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2014146707

Country of ref document: RU

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2013752676

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2013752676

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014032916

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112014032916

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20141229